WO2009020642A1 - Pyridine carboxamide orexin receptor antagonists - Google Patents

Pyridine carboxamide orexin receptor antagonists Download PDF

Info

Publication number
WO2009020642A1
WO2009020642A1 PCT/US2008/009491 US2008009491W WO2009020642A1 WO 2009020642 A1 WO2009020642 A1 WO 2009020642A1 US 2008009491 W US2008009491 W US 2008009491W WO 2009020642 A1 WO2009020642 A1 WO 2009020642A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
substituted
unsubstituted
alkyl
phenyl
Prior art date
Application number
PCT/US2008/009491
Other languages
French (fr)
Inventor
Jeffrey M. Bergman
Paul J. Coleman
Mark E. Fraley
Swati P. Mercer
Thomas S. Reger
Anthony J. Roecker
Justin T. Steen
Original Assignee
Merck & Co., Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merck & Co., Inc. filed Critical Merck & Co., Inc.
Priority to JP2010519985A priority Critical patent/JP2010535769A/en
Priority to CA2695621A priority patent/CA2695621A1/en
Priority to US12/669,909 priority patent/US8003797B2/en
Priority to EP08795115A priority patent/EP2184981A4/en
Priority to AU2008284268A priority patent/AU2008284268A1/en
Publication of WO2009020642A1 publication Critical patent/WO2009020642A1/en
Priority to US13/197,800 priority patent/US20110301140A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • C07D213/82Amides; Imides in position 3
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/08Drugs for disorders of the alimentary tract or the digestive system for nausea, cinetosis or vertigo; Antiemetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/14Antitussive agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings

Definitions

  • the orexins (hypocretins) comprise two neuropeptides produced in the hypothalamus: the orexin A (OX-A) (a 33 amino acid peptide) and the orexin B (OX-B) (a 28 amino acid peptide) (Sakurai T. et al., Cell, 1998, 92, 573-585). Orexins are found to stimulate food consumption in rats suggesting a physiological role for these peptides as mediators in the central feedback mechanism that regulates feeding behaviour (Sakurai T. et al., Cell, 1998, 92, 573-585).
  • Orexins regulate states of sleep and wakefulness opening potentially novel therapeutic approaches for narcoleptic or insomniac patients (Chemelli R.M. et al., Cell, 1999, 98, 437-451). Orexins have also been indicated as playing a role in arousal, reward, learning and memory (Harris, et al., Trends Neurosci., 2006, 29 (10), 571-577). Two orexin receptors have been cloned and characterized in mammals. They belong to the super family of G-protein coupled receptors (Sakurai T.
  • the orexin- 1 receptor (OX or OXlR) is selective for OX-A and the orexin-2 receptor (0X2 or 0X2R) is capable to bind OX-A as well as OX-B.
  • OX 1 receptor the orexin-1 receptor
  • OX 2 receptor the orexin-2 receptor (0X2 or 0X2R) is capable to bind OX-A as well as OX-B.
  • the physiological actions in which orexins are presumed to participate are thought to be expressed via one or both of OX 1 receptor and OX 2 receptor as the two subtypes of orexin receptors.
  • Orexin receptors are found in the mammalian brain and may have numerous implications in pathologies such as depression; anxiety; addictions; obsessive compulsive disorder; affective neurosis; depressive neurosis; anxiety neurosis; dysthymic disorder; behaviour disorder; mood disorder; sexual dysfunction; psychosexual dysfunction; sex disorder; schizophrenia; manic depression; delirium; dementia; severe mental retardation and dyskinesias such as Huntington's disease and Tourette syndrome; eating disorders such as anorexia, bulimia, cachexia, dysregulated appetite control; obesity; addictive feeding behaviors; binge/purge feeding behaviors; cardiovascular diseases; diabetes; appetite, taste, eating or drinking disorders; emesis, vomiting, nausea; asthma; cancer; Parkinson's disease; Cushing's syndrome/disease; basophile adenoma; prolactinoma; hyperprolactinemia; hypophysis tumour/adenoma; hypothalamic diseases; inflammatory bowel disease; gastric diskinesia; gastric ulcers
  • HIV post-chemotherapy pain
  • post-stroke pain post-operative pain
  • neuralgia conditions associated with visceral pain such as irritable bowel syndrome, and angina
  • urinary bladder incontinence e.g. urge incontinence
  • tolerance to narcotics or withdrawal from narcotics sleep disorders; sleep apnea; narcolepsy; insomnia; parasomnia; jet lag syndrome; and neurodegenerative disorders including nosological entities such as disinhibition-dementia-parkinsonism-amyotrophy complex; pallido-ponto-nigral degeneration; epilepsy; seizure disorders and other diseases related to general orexin system dysfunction.
  • Certain orexin receptor antagonists are disclosed in PCT patent publications WO
  • the present invention is directed to pyridyl carboxamide compounds which are antagonists of orexin receptors, and which are useful in the treatment or prevention of neurological and psychiatric disorders and diseases in which orexin receptors are involved.
  • the invention is also directed to pharmaceutical compositions comprising these compounds and the use of these compounds and compositions in the prevention or treatment of such diseases in which orexin receptors are involved.
  • the present invention is directed to compounds of the formula I:
  • A1 is selected from the group consisting of phenyl, naphthyl and heteroaryl
  • a 2 is selected from the group consisting of phenyl, naphthyl and heteroaryl
  • a 3 is selected from the group consisting of phenyl, naphthyl, C 3-6 cycloalkyl, and heterocycle;
  • R 1 a , R 1b and R 1 c may be absent if the valency of A 1 does not permit such substitution and are independently selected from the group consisting of:
  • R 2a , R 2b and R 2c may be absent if the valency of A 2 does not permit such substitution and are independently selected from the group consisting of:
  • R 3a , R 3b and R 3c may be absent if the valency of A ⁇ does not permit such substitution and are independently selected from the group consisting of:
  • R 4 and R 5 are independently selected from hydrogen and C 1 -6 alkyl, which is unsubstituted or substituted with one or more substituents selected from R 13 , or R 4 and R 5 may be joined together to form a C 3-6 cycloalkyl with the carbon atom to which they are attached, where the cycloalkyl is unsubstituted or substituted with one or more substituents selected from R 13 ;
  • R 6 is hydrogen, C 1 -6 alkyl or C 3-6 cycloalkyl, which is unsubstituted or substituted with one or more substituents selected from R 13 ;
  • R13 is selected from the group consisting of: (1) halogen,
  • R14 is selected from the group consisting of:
  • a 1 , A 2 , A 3 , R 1 a , R 1 b , RI C, R 2a , R 2b , R2C, R 3a , R 3b , R3C and R 4 are defined herein; or a pharmaceutically acceptable salt thereof.
  • a 1 , A 2 , A 3 , RIa, R 1 b , R 1 c ; R 2a , R 2b , R2C, R 3a , R 3b and R 3c are defined herein; or a pharmaceutically acceptable salt thereof.
  • An embodiment of the present invention includes compounds of the formula Ic:
  • a 2 , A 3 , R Ia, R 1 b , R Ic, R 2a , R 2b , R2C, R 3a , R 3b and R 3c are defined herein; or a pharmaceutically acceptable salt thereof.
  • An embodiment of the present invention includes compounds of the formula Ic':
  • a 2 , A 3 , R Ia, R 1 b , R 2a ; R 2b , R2C, R 3a , R 3b an d R 3c are defined herein; or a pharmaceutically acceptable salt thereof.
  • a 2 , A 3 , R 1 a , R 1 b , R 1 c , R 2a , R 2b , R2C, R 3a , R 3b and R 3c are defined herein; or a pharmaceutically acceptable salt thereof.
  • An embodiment of the present invention includes compounds of the formula Id':
  • a 2 , A 3 , R 1 a , R 1 b , R 1 c , R 2a , R 2b , R2C, R 3a , R 3b and R 3c are defined herein; or a pharmaceutically acceptable salt thereof.
  • a 3 , R 1 a , R 1 b , RI C, R 2a , R 2b , R2C, R 3a , R 3b and R 3c are defined herein; or a pharmaceutically acceptable salt thereof.
  • An embodiment of the present invention includes compounds of the formula If: wherein A 3 , R 1 a , RIb 3 RI C, R.2a, R 2b , R2C, R 3a , R 3b and R 3c are defined herein; or a pharmaceutically acceptable salt thereof.
  • a 2 , R 1 a , R 1 b , RI C, R 2a ; R 2b , R2C, R 3a , R 3b and R 3c are defined herein; or a pharmaceutically acceptable salt thereof.
  • An embodiment of the present invention includes compounds of the formula Ih:
  • a 1 , A 2 , R Ia, R 1 b , R Ic, R 2a , R 2b , R2C, R 3a , R 3b an d R 3c are defined herein; or a pharmaceutically acceptable salt thereof.
  • a 1 , A 2 , R 1 a , R 1 b ; RI C, R 2a , R 2b , R2C, R 3a , R 3b an d R 3c are defined herein; or a pharmaceutically acceptable salt thereof.
  • An embodiment of the present invention includes compounds of the formula Ij:
  • a 1 , A 2 , R 1 a , R 1 b , R 1 c , R.2a, R 2b , R2C, R 3a and R 3b are defined herein; or a pharmaceutically acceptable salt thereof.
  • a 1 , A 2 , R 1 a , R 1 b , R 1 c , R 2a , R 2b , R2C, R 3a and R 3b are defined herein; or a pharmaceutically acceptable salt thereof.
  • An embodiment of the present invention includes compounds of the formula II:
  • a 1 , A 2 , R 1 a , R 1 b , R 1 c , R 2a , R 2b and R 2c are defined herein; or a pharmaceutically acceptable salt thereof.
  • a 1 , A 2 , R 1 a , R 1 b ; R 1 c , R 2a , R 2b and R 2c are defined herein; or a pharmaceutically acceptable salt thereof.
  • An embodiment of the present invention includes compounds wherein A 1 is phenyl.
  • An embodiment of the present invention includes compounds wherein A 1 is naphthyl.
  • An embodiment of the present invention includes compounds wherein A 1 is heteroaryl.
  • An embodiment of the present invention includes compounds wherein A 1 is pyridyl.
  • An embodiment of the present invention includes compounds wherein A 2 is phenyl.
  • An embodiment of the present invention includes compounds wherein A 2 is naphthyl.
  • An embodiment of the present invention includes compounds wherein A 2 is heteroaryl.
  • An embodiment of the present invention includes compounds wherein A 2 is quinoxalinyl.
  • An embodiment of the present invention includes compounds wherein A 2 is pyridyl.
  • An embodiment of the present invention includes compounds wherein A 2 is pyrazinly.
  • An embodiment of the present invention includes compounds wherein A 2 is quinolinyl.
  • An embodiment of the present invention includes compounds wherein A 2 is indolyl.
  • An embodiment of the present invention includes compounds wherein A 2 is dihydroindolyl.
  • An embodiment of the present invention includes compounds wherein A 2 is benzimidazolyl.
  • An embodiment of the present invention includes compounds wherein A 3 is pyrazolyl.
  • An embodiment of the present invention includes compounds wherein A 3 is pyrazolyl which is substituted with methyl.
  • An embodiment of the present invention includes compounds wherein A 3 is pyridyl, which is substituted with at least one substituent other than hydrogen.
  • An embodiment of the present invention includes compounds wherein A 3 is pyridyl, which is substituted with fluroro or chloro.
  • An embodiment of the present invention includes compounds wherein A 3 is phenyl.
  • An embodiment of the present invention includes compounds wherein A 3 is phenyl, which is substituted with at least one substituent other than hydrogen.
  • An embodiment of the present invention includes compounds wherein A 3 is phenyl, which is substituted with hydroxyl, methylaminocarbonyl or dimethylaminomethyl.
  • An embodiment of the present invention includes compounds wherein A 3 is cyclopentyl.
  • An embodiment of the present invention includes compounds wherein A 3 is morpholinyl.
  • An embodiment of the present invention includes compounds wherein A 3 is pyrrolidinyl.
  • An embodiment of the present invention includes compounds wherein A 3 is azetidinyl.
  • R1 a , R 1 b and R1 c are independently selected from the group consisting of:
  • heteroaryl wherein heteroaryl is selected from pyrrolyl, imidazolyl, indolyl, pyridyl, and pyrimidinyl, which is unsubstituted or substituted with halogen, hydroxyl, C 1 -6 alkyl, -O-C 1 -6 alkyl or-NO2,
  • phenyl which is unsubstituted or substituted with halogen, hydroxyl, C 1 -6 alkyl, -O-C 1 -6 alkyl or-NO2,
  • R 1 a , R 1 b and R1 c are independently selected from the group consisting of:
  • R 1 a , R1 b and R1 c are independently selected from the group consisting of:
  • R 1 a , R 1 b and R1 c are independently selected from the group consisting of:
  • An embodiment of the present invention includes compounds wherein A 1 is phenyl and R 1 a , R 1 b and R1 c are independently selected from the group consisting of:
  • R 2a , R 2b and R 2c are independently selected from the group consisting of:
  • -O-C 1 -6 alkyl which is unsubstituted or substituted with halogen, hydroxyl or phenyl, (6) heteroaryl, wherein heteroaryl is selected from pyrrolyl, imidazolyl, indolyl, pyridyl, and pyrimidinyl, which is unsubstituted or substituted with halogen, hydroxyl, C 1 -6 alkyl, -O-C 1 -6 alkyl or-NO2,
  • phenyl which is unsubstituted or substituted with halogen, hydroxyl, C 1 -6 alkyl, -O-C 1 -6 alkyl or-NO2,
  • R 2a ; R 2b and R 2c are independently selected from the group consisting of:
  • R 2a , R 2b and R 2c are independently selected from the group consisting of:
  • An embodiment of the present invention includes compounds wherein R 2a ; R 2b and R 2c are independently selected from the group consisting of: (1) hydrogen,
  • An embodiment of the present invention includes compounds wherein A 2 is phenyl and R2 a , R 2b and R 2c are independently selected from the group consisting of:
  • An embodiment of the present invention includes compounds wherein R 3a , R 3b and R 3c are independently selected from the group consisting of: (1) hydrogen,
  • heteroaryl wherein heteroaryl is selected from pyrrolyl, imidazolyl, indolyl, pyridyl, and pyrimidinyl, which is unsubstituted or substituted with halogen, hydroxyl, Ci- ⁇ alkyl, -O-C 1 -6 alkyl or-NO2, (7) phenyl, which is unsubstituted or substituted with halogen, hydroxyl, C 1 -6 alkyl,
  • R 3a ; R.3b and R 3c are independently selected from the group consisting of:
  • R? a , R 3b and R 3c are independently selected from the group consisting of:
  • R 3a ; R 3b and R 3c are independently selected from the group consisting of:
  • An embodiment of the present invention includes compounds wherein A 3 is pyridyl and wherein R 3a j s halogen, R 3b is hydrogen and R 3c is hydrogen.
  • An embodiment of the present invention includes compounds wherein A 3 is pyridyl and wherein R 3a is chloro or fluoro, R 3b is hydrogen and R 3c is hydrogen.
  • An embodiment of the present invention includes compounds wherein A 3 is pyrazolyl and wherein R 3a is C 1 -6 alkyl, R 3b is hydrogen and R 3c is hydrogen.
  • An embodiment of the present invention includes compounds wherein A 3 is pyrazolyl and wherein R 3a is methyl, R 3b is hydrogen and R 3c is hydrogen.
  • An embodiment of the present invention includes compounds wherein R 4 is hydrogen or C 1 -6 alkyl.
  • An embodiment of the present invention includes compounds wherein
  • R 4 is hydrogen or methyl.
  • An embodiment of the present invention includes compounds wherein
  • R 4 is hydrogen.
  • An embodiment of the present invention includes compounds wherein R ⁇ is hydrogen or C 1 -6 alkyl.
  • An embodiment of the present invention includes compounds wherein
  • R 5 is hydrogen or methyl.
  • An embodiment of the present invention includes compounds wherein
  • R 5 is hydrogen
  • An embodiment of the present invention includes compounds wherein R 6 is hydrogen, C 1 -6 alkyl or C 3-6 cycloalkyl. An embodiment of the present invention includes compounds wherein R 6 is C 1 -6 alkyl. An embodiment of the present invention includes compounds wherein R 6 is C 3-6 cycloalkyl. An embodiment of the present invention includes compounds wherein R 6 is hydrogen, methyl or ethyl. An embodiment of the present invention includes compounds wherein R 6 is hydrogen.
  • Specific embodiments of the present invention include a compound which is selected from the group consisting of the subject compounds of the Examples herein or a pharmaceutically acceptable salt thereof.
  • the compounds of the present invention may contain one or more asymmetric centers and can thus occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. Additional asymmetric centers may be present depending upon the nature of the various substituents on the molecule. Each such asymmetric center will independently produce two optical isomers and it is intended that all of the possible optical isomers and diastereomers in mixtures and as pure or partially purified compounds are included within the ambit of this invention. The present invention is meant to comprehend all such isomeric forms of these compounds.
  • Formula I shows the structure of the class of compounds without specific stereochemistry.
  • the coupling reaction is often the formation of salts using an enantiomerically pure acid or base.
  • the diasteromeric derivatives may then be converted to the pure enantiomers by cleavage of the added chiral residue.
  • the racemic mixture of the compounds can also be separated directly by chromatographic methods utilizing chiral stationary phases, which methods are well known in the art.
  • a 1 ternatively, any enantiomer of a compound may be obtained by stereoselective synthesis using optically pure starting materials or reagents of known configuration by methods well known in the art.
  • halogen or halo as used herein are intended to include fluoro, chloro, bromo and iodo.
  • C 1 -6 as in C 1 -6 alkyl is defined to identify the group as having 1, 2, 3, 4, 5 or 6 carbons in a linear or branched arrangement, such that Ci-8alkyl specifically includes methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert- butyl, pentyl, and hexyl.
  • a group which is designated as being independently substituted with substituents may be independently substituted with multiple numbers of such substituents.
  • heterocycle includes both unsaturated and saturated heterocyclic moieties, wherein the unsaturated heterocyclic moieties (i.e. "heteroaryl”) include benzoimidazolyl, benzimidazolonyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzothiazolyl, benzotriazolyl, benzothiophenyl, benzoxazepin, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, imidazolyl, indolinyl, indolyl, dihydroindolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthpyridinyl, oxadiazolyl, oxazolyl, oxazoline,
  • salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc, and the like. Particular embodiments include the ammonium, calcium, magnesium, potassium, and sodium salts. Salts in the solid form may exist in more than one crystal structure, and may also be in the form of hydrates.
  • Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N'-dibenzylethylene-diamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethyl-morpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like.
  • basic ion exchange resins such
  • salts may be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids.
  • acids include acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p- toluenesulfonic acid, and the like.
  • Particular embodiments include the citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, fumaric, and tartaric acids. It will be understood that, as used herein, references to the compounds of Formula I are meant to also include the pharmaceutically acceptable salts.
  • Exemplifying the invention is the use of the compounds disclosed in the Examples and herein.
  • Specific compounds within the present invention include a compound which selected from the group consisting of the compounds disclosed in the following Examples and pharmaceutically acceptable salts thereof and individual enantiomers or diastereomers thereof.
  • the subject compounds are useful in a method of antagonizing orexin receptor activity in a patient such as a mammal in need of such inhibition comprising the administration of an effective amount of the compound.
  • the present invention is directed to the use of the compounds disclosed herein as antagonists of orexin receptor activity. In addition to primates, especially humans, a variety of other mammals can be treated according to the method of the present invention.
  • the present invention is directed to a compound of the present invention or a pharmaceutically acceptable salt thereof for use in medicine.
  • the present invention is further directed to a use of a compound of the present invention or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for antagonizing orexin receptor activity or treating the disorders and diseases noted herein in humans and animals.
  • the subject treated in the present methods is generally a mammal, such as a human being, male or female.
  • the term "therapeutically effective amount” means the amount of the subject compound that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician. It is recognized that one skilled in the art may affect the neurological and psychiatric disorders by treating a patient presently afflicted with the disorders or by prophylactically treating a patient afflicted with the disorders with an effective amount of the compound of the present invention.
  • treatment and “treating” refer to all processes wherein there may be a slowing, interrupting, arresting, controlling, or stopping of the progression of the neurological and psychiatric disorders described herein, but does not necessarily indicate a total elimination of all disorder symptoms, as well as the prophylactic therapy of the mentioned conditions, particularly in a patient who is predisposed to such disease or disorder.
  • administration of and or “administering a” compound should be understood to mean providing a compound of the invention or a prodrug of a compound of the invention to the individual in need thereof.
  • composition as used herein is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
  • Such term in relation to pharmaceutical composition is intended to encompass a product comprising the active ingredient(s), and the inert ingredient(s) that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients.
  • the pharmaceutical compositions of the present invention encompass any composition made by admixing a compound of the present invention and a pharmaceutically acceptable carrier.
  • the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • pharmaceutically acceptable it is meant the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • the utility of the compounds in accordance with the present invention as orexin receptor OXlR and/or 0X2R antagonists may be readily determined without undue experimentation by methodology well known in the art, including the “FLIPR Ca 2+ Flux Assay” (Okumura et al., Biochem. Biophys. Res. Comm. 280:976-981, 2001). In a typical experiment the OXl and 0X2 receptor antagonistic activity of the compounds of the present invention was determined in accordance with the following experimental method.
  • CHO cells expressing the rat orexin- 1 receptor or the human orexin-2 receptor, are grown in Iscove's modified DMEM containing 2 mM L-glutamine, 0.5 g/ml G418, 1% hypoxanthine-thymidine supplement, 100 U/ml penicillin, 100 ug/ml streptomycin and 10 % heat-inactivated fetal calf serum (FCS).
  • FCS heat-inactivated fetal calf serum
  • the cells are seeded at 20,000 cells / well into Becton-Dickinson black 384-well clear bottom sterile plates coated with poly-D- lysine. All reagents were from GIBCO-Invitrogen Corp.
  • Ala-6,12 human orexin-A as the agonist is prepared as a 1 mM stock solution in 1% bovine serum albumin (BSA) and diluted in assay buffer (HBSS containing 20 mM HEPES, 0.1% BSA and 2.5mM probenecid, pH7.4) for use in the assay at a final concentration of 7OpM.
  • Test compounds are prepared as 10 mM stock solution in DMSO, then diluted in 384-well plates, first in DMSO, then assay buffer.
  • test compounds are added to the plate in a volume of 25 ul, incubated for 5 min and finally 25 ul of agonist is added. Fluorescence is measured for each well at 1 second intervals for 5 minutes and the height of each fluorescence peak is compared to the height of the fluorescence peak induced by 70 pM Ala-6,12 orexin-A with buffer in place of antagonist. For each antagonist, IC50 value (the concentration of compound needed to inhibit 50 % of the agonist response) is determined. A 1 ternatively, compound potency can be assessed by a radioligand binding assay (described in Bergman et. al. Bioorg. Med. Chem. Lett.
  • the inhibition constant (Kj) is determined in membranes prepared from CHO cells expressing either the OXl or OX2 receptor.
  • the intrinsic orexin receptor antagonist activity of a compound which may be used in the present invention may be determined by these assays.
  • the compounds of the following examples had activity in antagonizing the rat orexin-1 receptor and/or the human orexin-2 receptor in the aforementioned assays, generally with an IC50 of less than about 50 ⁇ M.
  • Many of compounds within the present invention had activity in antagonizing the rat orexin-1 receptor and/or the human orexin-2 receptor in the aforementioned assays with an IC50 of less than about 100 nM.
  • Compounds of the present invention also have activity in the radioligand binding assay, generally with a Ki ⁇ 100 nM against the orexin-1 and/or the orexin-2 receptor. Additional data is provided in the following Examples. Such a result is indicative of the intrinsic activity of the compounds in use as antagonists of orexin-1 receptor and/or the orexin-2 receptor.
  • the present invention also includes compounds within the generic scope of the invention which possess activity as agonists of the orexin-1 receptor and/or the orexin-2 receptor.
  • the present compounds exhibit unexpected properties, such as with respect to increased oral bioavailability, metabolic stability, decreased inhibition of metabolic enzymes (such as decreased cytochrome P450 3A4 (CYP3A4) inhibition), decreased inhibition of transporters (such as decreased p-glycoprotein/PGP inhibition) and/or selectivity with respect to other receptors, including the human orexin-2 receptor.
  • metabolic enzymes such as decreased cytochrome P450 3A4 (CYP3A4) inhibition
  • transporters such as decreased p-glycoprotein/PGP inhibition
  • selectivity with respect to other receptors, including the human orexin-2 receptor.
  • the orexin receptors have been implicated in a wide range of biological functions. This has suggested a potential role for these receptors in a variety of disease processes in humans or other species.
  • the compounds of the present invention have utility in treating, preventing, ameliorating, controlling or reducing the risk of a variety of neurological and psychiatric disorders associated with orexin receptors, including one or more of the following conditions or diseases: sleep disorders, sleep disturbances, including enhancing sleep quality, improving sleep quality, increasing sleep efficiency, augmenting sleep maintenance; increasing the value which is calculated from the time that a subject sleeps divided by the time that a subject is attempting to sleep; improving sleep initiation; decreasing sleep latency or onset (the time it takes to fall asleep); decreasing difficulties in falling asleep; increasing sleep continuity; decreasing the number of awakenings during sleep; decreasing intermittent wakings during sleep; decreasing nocturnal arousals; decreasing the time spent awake following the initial onset of sleep; increasing the total amount of sleep; reducing the fragmentation of sleep; altering the timing, frequency or duration of REM sleep bout
  • HIV post-chemotherapy pain; post-stroke pain; post-operative pain; neuralgia; emesis, nausea, vomiting; gastric dyskinesia; gastric ulcers; Kallman's syndrome (anosmia); conditions associated with visceral pain such as irritable bowel syndrome, and angina; eating disorders; urinary incontinence; substance tolerance, substance withdrawal (including, substances such as opiates, nicotine, tobacco products, alcohol, benzodiazepines, cocaine, sedatives, hypnotics, etc.); psychosis; schizophrenia; anxiety (including generalized anxiety disorder, panic disorder, and obsessive compulsive disorder); mood disorders (including depression, mania, bipolar disorders); trigeminal neuralgia; hearing loss; tinnitus; neuronal damage including ocular damage; retinopathy; macular degeneration of the eye; emesis; brain edema; pain, including acute and chronic pain states, severe pain, intractable pain, inflammatory pain, neuropathic pain
  • the present invention provides methods for: enhancing the quality of sleep; augmenting sleep maintenance; increasing REM sleep; increasing stage 2 sleep; decreasing fragmentation of sleep patterns; treating insomnia and all types of sleep disorders; treating or controlling sleep disturbances associated with diseases such as neurological disorders including neuropathic pain and restless leg syndrome; treating or controlling addiction disorders; treating or controlling psychoactive substance use and abuse; enhancing cognition; increasing memory retention; treating or controlling obesity; treating or controlling diabetes and appetite, taste, eating, or drinking disorders; treating or controlling hypothalamic diseases; treating or controlling depression; treating, controlling, ameliorating or reducing the risk of epilepsy, including absence epilepsy; treating or controlling pain, including neuropathic pain; treating or controlling Parkinson's disease; treating or controlling psychosis; treating or controlling dysthymic, mood, psychotic and anxiety disorders; treating or controlling depression, including major depression and major dperession disorder; treating or controlling bipolar disorder; or treating, controlling, ameliorating or reducing the risk of schizophrenia, in a mammalian patient in need thereof which comprises administering
  • the subject compounds are further useful in a method for the prevention, treatment, control, amelioration, or reducation of risk of the diseases, disorders and conditions noted herein.
  • the dosage of active ingredient in the compositions of this invention may be varied, however, it is necessary that the amount of the active ingredient be such that a suitable dosage form is obtained.
  • the active ingredient may be administered to patients (animals and human) in need of such treatment in dosages that will provide optimal pharmaceutical efficacy.
  • the selected dosage depends upon the desired therapeutic effect, on the route of administration, and on the duration of the treatment.
  • the dose will vary from patient to patient depending upon the nature and severity of disease, the patient's weight, special diets then being followed by a patient, concurrent medication, and other factors which those skilled in the art will recognize.
  • dosage levels of between 0.0001 to 10 mg/kg. of body weight daily are administered to the patient, e.g., humans and elderly humans, to obtain effective antagonism of orexin receptors.
  • the dosage range will generally be about 0.5 mg to 1.0 g. per patient per day which may be administered in single or multiple doses. In one embodiment, the dosage range will be about 0.5 mg to 500 mg per patient per day; in another embodiment about 0.5 mg to 200 mg per patient per day; and in yet another embodiment about 5 mg to 50 mg per patient per day.
  • Pharmaceutical compositions of the present invention may be provided in a solid dosage formulation such as comprising about 0.5 mg to 500 mg active ingredient, or comprising about 1 mg to 250 mg active ingredient.
  • the pharmaceutical composition may be provided in a solid dosage formulation comprising about 1 mg, 5 mg, 10 mg, 25 mg, 30 mg, 50 mg, 80 mg, 100 mg, 200 mg or 250 mg active ingredient.
  • the compositions may be provided in the form of tablets containing 1.0 to 1000 milligrams of the active ingredient, such as 1, 5, 10, 15, 20, 25, 50, 75, 100, 150, 200, 250, 300, 400, 500, 600, 750, 800, 900, and 1000 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated.
  • the compounds may be administered on a regimen of 1 to 4 times per day, such as once or twice per day.
  • the compounds may be administered before bedtime. For example, the compounds may be administered about 1Hour prior to bedtime, about 30 minutes prior to bedtime or immediately before bedtime.
  • the compounds of the present invention may be used in combination with one or more other drugs in the treatment, prevention, control, amelioration, or reduction of risk of diseases or conditions for which compounds of the present invention or the other drugs may have utility, where the combination of the drugs together are safer or more effective than either drug alone.
  • Such other drug(s) may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with a compound of the present invention.
  • a pharmaceutical composition in unit dosage form containing such other drugs and the compound of the present invention is contemplated.
  • the combination therapy may also includes therapies in which the compound of the present invention and one or more other drugs are administered on different overlapping schedules.
  • the compounds of the present invention and the other active ingredients may be used in lower doses than when each is used singly.
  • the pharmaceutical compositions of the present invention include those that contain one or more other active ingredients, in addition to a compound of the present invention.
  • the above combinations include combinations of a compound of the present invention not only with one other active compound, but also with two or more other active compounds.
  • compounds of the present invention may be used in combination with other drugs that are used in the prevention, treatment, control, amelioration, or reduction of risk of the diseases or conditions for which compounds of the present invention are useful.
  • Such other drugs may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with a compound of the present invention.
  • a pharmaceutical composition containing such other drugs in addition to the compound of the present invention is contemplated.
  • the pharmaceutical compositions of the present invention include those that also contain one or more other active ingredients, in addition to a compound of the present invention.
  • the weight ratio of the compound of the compound of the present invention to the second active ingredient may be varied and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used. Thus, for example, when a compound of the present invention is combined with another agent, the weight ratio of the compound of the present invention to the other agent will generally range from about 1000:1 to about 1 :1000, such as about 200:1 to about 1 :200. Combinations of a compound of the present invention and other active ingredients will generally also be within the aforementioned range, but in each case, an effective dose of each active ingredient should be used. In such combinations the compound of the present invention and other active agents may be administered separately or in conjunction.
  • the administration of one element may be prior to, concurrent to, or subsequent to the administration of other agent(s).
  • the compounds of the present invention may be administered in conbination with other compounds which are known in the art to be useful for enhancing sleep quality and preventing and treating sleep disorders and sleep disturbances, including e.g., sedatives, hypnotics, anxiolytics, antipsychotics, antianxiety agents, antihistamines, benzodiazepines, barbiturates, cyclopyrrolones, GABA agonists, 5HT-2 antagonists including 5HT-2A antagonists and 5HT-2A/2C antagonists, histamine antagonists including histamine H3 antagonists, histamine H3 inverse agonists, imidazopyridines, minor tranquilizers, melatonin agonists and antagonists, melatonergic agents, other orexin antagonists, orexin agonists, prokineticin agonists and antagonists, pyrazolopyrimidines, T-type calcium channel antagonists
  • the subject compound may be employed in combination with other compounds which are known in the art, either administered separately or in the same pharmaceutical compositions, include, but are not limited to: insulin sensitizers including (i) PPAR ⁇ antagonists such as glitazones (e.g.
  • ciglitazone darglitazone; englitazone; isaglitazone (MCC-555); pioglitazone; rosiglitazone; troglitazone; tularik; BRL49653; CLX-0921; 5-BTZD), GW-0207, LG- 100641, and LY-300512, and the like);
  • biguanides such as metformin and phenformin
  • insulin or insulin mimetics such as biota, LP-100, novarapid, insulin detemir, insulin lispro, insulin glargine, insulin zinc suspension (lente and ultralente); Lys-Pro insulin, GLP-I (73-7) (insulintropin); and GLP-I (7-36)-NH2)
  • sulfonylureas such as acetohexamide; chlorpropamide; diabinese; glibenclamide; glipizide; gly
  • PTP-IB protein tyrosine phosphatase- IB
  • cannabinoid receptor ligands such as cannabinoid CBi receptor antagonists or inverse agonists, such as rimonabant, taranabant
  • WO 01/77094 (7) neuropeptide Yl antagonists, such as BIBP3226, J-1 15814, BIBO 3304, LY- 357897, CP-671906, GI-264879A, and those disclosed in U.S. Patent No. 6,001,836, and PCT Patent Publication Nos.
  • WO 96/14307 WO 01/23387, WO 99/51600, WO 01/85690, WO 01/85098, WO 01/85173, and WO 01/89528;
  • neuropeptide Y5 antagonists such as GW- 569180A, GW-594884A, GW-587081X, GW-5481 18X, FR226928, FR 240662, FR252384, 1229U91 , GI-264879A, CGP71683A, LY-377897, PD-160170, SR-120562A, SR-120819A and JCF-104, and those disclosed in U.S. Patent Nos.
  • WO 97/19682 WO 97/20820, WO 97/20821, WO 97/20822, WO 97/20823, WO 98/24768; WO 98/25907; WO 98/25908; WO 98/27063, WO 98/47505; WO 98/40356; WO 99/15516; WO 99/27965; WO 00/64880, WO 00/68197, WO 00/69849, WO 01/09120, WO 01/14376; WO 01/85714, WO 01/85730, WO 01/07409, WO 01/02379, WO 01/02379, WO 01/23388, WO 01/23389, WO 01/44201, WO 01/62737, WO 01/62738, WO 01/09120, WO 02/22592, WO 0248152, and WO 02/49648; WO 02/094825; WO 03/0140
  • GLP-I agonists such as GLP-I agonists; (22) corticotropin-releasing hormone agonists; (23) histamine receptor-3 (H3) modulators; (24) histamine receptor-3 (H3) antagonists/inverse agonists, such as hioperamide, 3- (1H-imidazol-4-yl)propyl N-(4-pentenyl)carbamate, clobenpropit, iodophenpropit, imoproxifan, GT2394 (Gliatech), and O-[3-(1H-imidazol-4-yl)propanol]-carbamates; (25) ⁇ -hydroxy steroid dehydrogenase- 1 inhibitors ( ⁇ -HSD-1); (26) PDE (phosphodiesterase) inhibitors, such as theophylline, pentoxifylline, zaprinast, sildenafil, amrinone, milrinone, cilostamide, rolipram, and cilomilast
  • leptin including recombinant human leptin (PEG-OB, Hoffman La Roche) and recombinant methionyl human leptin (Amgen); (31) leptin derivatives; (32) BRS3 (bombesin receptor subtype 3) agonists. such as [D-Phe6,beta-Alal l,Phel 3,Nlel4]Bn(6-14) and [D-
  • CNTF Central neurotrophic factors
  • GI-181771 Gaxo-SmithKline
  • SR 146131 Sanofi Synthelabo
  • butabindide PD 170,292, and PD 149164 (Pfizer)
  • CNTF derivatives such as axokine (Regeneron)
  • monoamine reuptake inhibitors such as sibutramine
  • UCP-I uncoupling protein-1
  • activators such as phytanic acid, 4-[(E)- 2-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-napthalenyl)-1-propenyl]benzoic acid (TTNPB), retinoic acid
  • thyroid hormone ⁇ agonists such as KB-261 1 (K
  • dipeptidyl peptidase IV (DP-IV) inhibitors such as isoleucine thiazolidide, valine pyrrolidide, NVP-DPP728, LAF237, P93/01, TSL 225, TMC-2A/2B/2C, FE 99901 1, P9310/K364, VIP 0177, SDZ 274-444, sitagliptin; and the compounds disclosed in US 6,699,871 , WO 03/004498; WO 03/004496; EP 1 258 476; WO 02/083128; WO 02/062764; WO 03/000250; WO 03/002530; WO 03/002531 ; WO 03/002553; WO 03/002593; WO 03/000180; and WO 03/000181; (46) dicarboxylate transporter inhibitors; (47) glucose transporter inhibitors; (48)
  • Neuropeptide Y2 (NPY2) receptor agonists such NPY3-36, N acetyl [Leu(28,31)] NPY 24-36, TASP-V, and cyclo-(28/32)-Ac-[Lys28-Glu32]-(25-36)-pNPY;
  • Neuropeptide Y4 (NP Y4) agonists such as pancreatic peptide (PP), and other Y4 agonists such as 1229U91;
  • cyclooxygenase-2 inhibitors such as etoricoxib, celecoxib, valdecoxib, parecoxib, lumiracoxib, BMS347070, tiracoxib or JTE522, ABT963, CS502 and GW406381 ;
  • NPY2 (NPY2) receptor agonists such NPY3-36, N acetyl [Leu(28,31)] NPY 24-36, TASP-V, and cyclo-(28
  • Neuropeptide Yl (NPYl) antagonists such as BIBP3226, J-115814, BIBO 3304, LY-357897, CP-671906, GI-264879A; (56) Opioid antagonists such as nalmefene (Revex ®), 3- methoxynaltrexone, naloxone, naltrexone; (57) 1 l ⁇ HSD-I (11 -beta hydroxy steroid dehydrogenase type 1) inhibitors such as BVT 3498, BVT 2733, and those disclosed in WO 01/90091, WO 01/90090, WO 01/90092, US 6,730,690 and US 2004-013301 1; (58) aminorex; (59) amphechloral; (60) amphetamine; (61) benzphetamine; (62) chlorphentermine; (63) clobenzorex; (64) cloforex; (65) clominorex; (66) c
  • the subject compound may be employed in combination with an anti-depressant or anti-anxiety agent, including norepinephrine reuptake inhibitors (including tertiary amine tricyclics and secondary amine tricyclics), selective serotonin reuptake inhibitors (SSRIs), monoamine oxidase inhibitors (MAOIs), reversible inhibitors of monoamine oxidase (RIMAs), serotonin and noradrenaline reuptake inhibitors (SNRIs), corticotropin releasing factor (CRF) antagonists, ⁇ -adrenoreceptor antagonists, neurokinin- 1 receptor antagonists, atypical anti-depressants, benzodiazepines, 5-HT I A agonists or antagonists, especially 5-
  • Specific agents include: amitriptyline, clomipramine, doxepin, imipramine and trimipramine; amoxapine, desipramine, maprotiline, nortriptyline and protriptyline; citalopram, duloxetine, fluoxetine, fluvoxamine, paroxetine and sertraline; isocarboxazid, phenelzine, tranylcypromine and selegiline; moclobemide: venlafaxine; aprepitant; bupropion, lithium, nefazodone, trazodone and viloxazine; alprazolam, chlordiazepoxide, clonazepam, chlorazepate, diazepam, halazepam, lorazepam, oxazepam and prazepam; buspirone, flesinoxan, gepirone and ipsapirone, and pharmaceutically acceptable salts thereof.
  • the subject compound may be employed in combination with anti- Alzheimer's agents; beta-secretase inhibitors; gamma-secretase inhibitors; growth hormone secretagogues; recombinant growth hormone; HMG-CoA reductase inhibitors; NS AID's including ibuprofen; vitamin E; anti-amyloid antibodies; CB-I receptor antagonists or CB-I receptor inverse agonists; antibiotics such as doxycycline and rifampin; N-methyl-D- aspartate (NMDA) receptor antagonists, such as memantine; cholinesterase inhibitors such as galantamine, rivastigmine, donepezil, and tacrine; growth hormone secretagogues such as ibutamoren, ibutamoren mesylate, and capromorelin; histamine H3 antagonists; AMPA agonists; PDE IV inhibitors; GABAA inverse agonists; or neuronal nicotinic agonists
  • the subject compound may be employed in combination with sedatives, hypnotics, anxiolytics, antipsychotics, antianxiety agents, cyclopyrrolones, imidazopyridines, pyrazolopyrimidines, minor tranquilizers, melatonin agonists and antagonists, melatonergic agents, benzodiazepines, barbiturates, 5HT-2 antagonists, and the like, such as: adinazolam, allobarbital, alonimid, alprazolam, amitriptyline, amobarbital, amoxapine, bentazepam, benzoctamine, brotizolam, bupropion, busprione, butabarbital, butalbital, capuride, carbocloral, chloral betaine, chloral hydrate, chlordiazepoxide, clomipramine, clonazepam, cloperidone, clorazepate, clorethate
  • the subject compound may be employed in combination with levodopa (with or without a selective extracerebral decarboxylase inhibitor such as carbidopa or benserazide), anticholinergics such as biperiden (optionally as its hydrochloride or lactate salt) and trihexyphenidyl (benzhexol) hydrochloride, COMT inhibitors such as entacapone, MOA-B inhibitors, antioxidants, A2a adenosine receptor antagonists, cholinergic agonists, NMDA receptor antagonists, serotonin receptor antagonists and dopamine receptor agonists such as alentemol, bromocriptine, fenoldopam, lisuride, naxagolide, pergolide and pramipexole.
  • levodopa with or without a selective extracerebral decarboxylase inhibitor such as carbidopa or benserazide
  • anticholinergics such as biperi
  • the subject compound may be employed in combination with acetophenazine, alentemol, benzhexol, bromocriptine, biperiden, chlorpromazine, chlorprothixene, clozapine, diazepam, fenoldopam, fluphenazine, haloperidol, levodopa, levodopa with benserazide, levodopa with carbidopa, lisuride, loxapine, mesoridazine, molindolone, naxagolide, olanzapine, pergolide, perphenazine, pimozide, pramipexole, risperidone, sulpiride, tetrabenazine, trihexyphenidyl, thioridazine, thiothixene or trifluoperazine.
  • the subject compound may be employed in combination with a compound from the phenothiazine, thioxanthene, heterocyclic dibenzazepine, butyrophenone, diphenylbutylpiperidine and indolone classes of neuroleptic agent.
  • phenothiazines include chlorpromazine, mesoridazine, thioridazine, acetophenazine, fluphenazine, perphenazine and trifluoperazine.
  • Suitable examples of thioxanthenes include chlorprothixene and thiothixene.
  • An example of a dibenzazepine is clozapine.
  • An example of a butyrophenone is haloperidol.
  • An example of a diphenylbutylpiperidine is pimozide.
  • An example of an indolone is molindolone.
  • Other neuroleptic agents include loxapine, sulpiride and risperidone.
  • the subject compound may be employed in combination with a nicotine agonist or a nicotine receptor partial agonist such as varenicline, opioid antagonists (e.g., naltrexone (including naltrexone depot), antabuse, and nalmefene), dopaminergic agents (e.g., apomorphine), ADD/ ADHD agents (e.g., methylphenidate hydrochloride (e.g., R1talin® and Concerta®), atomoxetine (e.g., Strattera®), a monoamine oxidase inhibitor (MAOI), amphetamines (e.g., Adderall®)) and anti-obesity agents, such as apo- B/MTP inhibitors, 1 1 Beta-hydroxy steroid dehydrogenase- 1 (1 IBeta-HSD type 1) inhibitors, peptide YY3-36 or analogs thereof, MCR-4 agonists, CCK-A agonists, monoamine renicline,
  • the subject compound may be employed in combination with an anoretic agent such as aminorex, amphechloral, amphetamine, benzphetamine, chlorphentermine, clobenzorex, cloforex, clominorex, clortermine, cyclexedrine, dexfenfluramine, dextroamphetamine, diethylpropion, diphemethoxidine, N-ethylamphetamine, fenbutrazate, fenfluramine, fenisorex, fenproporex, fludorex, fluminorex, furfurylmethylamphetamine, levamfetamine, levophacetoperane, mazindol, mefenorex, metamfepramone, methamphetamine, norpseudoephedrine, pentorex, phendimetrazine, phenmetrazine, phentermine, phenylpropanolamine, pic
  • the subject compound may be employed in combination with an opiate agonist, a lipoxygenase inhibitor, such as an inhibitor of 5-lipoxygenase, a cyclooxygenase inhibitor, such as a cyclooxygenase-2 inhibitor, an interleukin inhibitor, such as an interleukin- 1 inhibitor, an NMDA antagonist, an inhibitor of nitric oxide or an inhibitor of the synthesis of nitric oxide, a non-steroidal antiinflammatory agent, or a cytokine-suppressing antiinflammatory agent, for example with a compound such as acetaminophen, asprin, codiene, fentanyl, ibuprofen, indomethacin, ketorolac, morphine, naproxen, phenacetin, piroxicam, a steroidal analgesic, sufentanyl, sunlindac, tenidap, and the like.
  • a lipoxygenase inhibitor such as an inhibitor of 5-lip
  • the subject compound may be administered with a pain reliever; a potentiator such as caffeine, an H2-antagonist, simethicone, aluminum or magnesium hydroxide; a decongestant such as phenylephrine, phenylpropanolamine, pseudophedrine, oxymetazoline, epinephrine, naphazoline, xylometazoline, propylhexedrine, or levo-desoxy-ephedrine; an antiitussive such as codeine, hydrocodone, caramiphen, carbetapentane, or dextramethorphan; a diuretic; and a sedating or non-sedating antihistamine.
  • a pain reliever such as caffeine, an H2-antagonist, simethicone, aluminum or magnesium hydroxide
  • a decongestant such as phenylephrine, phenylpropanolamine, pseudophedrine, oxymetazoline, epinep
  • the compounds of the present invention may be administered by oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, ICV, intracisternal injection or infusion, subcutaneous injection, or implant), by inhalation spray, nasal, vaginal, rectal, sublingual, or topical routes of administration and may be formulated, alone or together, in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles appropriate for each route of administration.
  • parenteral e.g., intramuscular, intraperitoneal, intravenous, ICV, intracisternal injection or infusion, subcutaneous injection, or implant
  • inhalation spray nasal, vaginal, rectal, sublingual, or topical routes of administration
  • nasal, vaginal, rectal, sublingual, or topical routes of administration may be formulated, alone or together, in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles appropriate for each route of administration.
  • the compounds of the invention are effective for
  • compositions for the administration of the compounds of this invention may conveniently be presented in dosage unit form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the active ingredient into association with the carrier which constitutes one or more accessory ingredients.
  • the pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation.
  • the active object compound is included in an amount sufficient to produce the desired effect upon the process or condition of diseases.
  • composition is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
  • Pharmaceutical compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • compositions for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
  • Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • Oily suspensions may be formulated by suspending the active ingredient in a suitable oil. Oil-in-water emulsions may also be employed.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • Pharmaceutical compositions of the present compounds may be in the form of a sterile injectable aqueous or oleagenous suspension.
  • the compounds of the present invention may also be administered in the form of suppositories for rectal administration.
  • creams, ointments, jellies, solutions or suspensions, etc., containing the compounds of the present invention may be employed.
  • the compounds of the present invention may also be formulated for administered by inhalation.
  • the compounds of the present invention may also be administered by a transdermal patch by methods known in the art.
  • the final product may be further modified, for example, by manipulation of substituents.
  • substituents may include, but are not limited to, reduction, oxidation, alkylation, acylation, and hydrolysis reactions which are commonly known to those skilled in the art.
  • the order of carrying out the foregoing reaction schemes may be varied to facilitate the reaction or to avoid unwanted reaction products.
  • the following examples are provided so that the invention might be more fully understood. These examples are illustrative only and should not be construed as limiting the invention in any way.
  • A-2 5-Aryl-2-chloronicotinic acids (A-2) are synthesized via Suzuki coupling reactions on A-I with aryl boronic under standard conditions followed by hydrolysis. A-2 are coupled to amines using PyCIu to give A-3. Coupling of A-3 with boronic acids using standard Suzuki coupling conditions under microwave heating affords A-4.
  • B-2 2,5-Disubstituted nicotinic acids
  • B-I 2,5-Disubstituted nicotinic acids
  • C-2 2,5-Disubstituted nicotinic acids
  • C-I 2,5-Disubstituted nicotinic acids
  • D-2 2,5-Disubstituted nicotinic acids
  • D-I 2,5-Disubstituted nicotinic acids
  • E-2 2,5-Disubstituted nicotinic acids
  • E-1 2,5-Disubstituted nicotinic acids
  • E-2 can be synthesized via E-1 by selective Suzuki coupling on the 5-position followed by nucleophilic substitution with N-H containing heterocycles on the 2-position. Hydrolysis of the ester affords E-2 Compounds such as E-2 can be advanced to final compounds through amide coupling to afford E-3.
  • F-2 2,5-Disubstituted nicotinic acids
  • F-2 can be synthesized via IM .
  • Hydrolysis of the ester affords F-2.
  • Compounds such as F-2 can be advanced to final compounds through amide coupling to afford F-3.
  • G-2 2,5-Disubstituted nicotinic esters
  • G-I 2,5-Disubstituted nicotinic esters
  • Compounds such as G-2 can be advanced to final compounds through transamination mediated by trimethylaluminum to afford G-3.
  • H-2 2,5-Disubstituted nicotinic acids
  • reaction mixture was partitioned between ethyl acetate and water, washed with saturated sodium carbonate and dried over magnesium sulfate.
  • the reaction mixture was partitioned between ethyl acetate and saturated sodium bicarbonate. The organics were washed with water and brine, then stripped to dryness.
  • the crude product was taken up in DMF/DMSO (30 mL, 1 :1), filtered and purified via reverse phase chromatography (5 ⁇ 65% acetonitrile in water with 0.1% TFA buffer). The clean fractions were pooled, partitioned between ethyl acetate and 2M sodium carbonate. The organic layer was washed with water and brine, dried over anhydrous sodium sulfate, filtered and concentrated to afford the title compound (3-5) as a white solid.
  • reaction mixture was cooled and partitioned between EtOAc and water, washed with saturated sodium bicarbonate and dried over magnesium sulfate.
  • the reaction mixture was filtered, concentrated and purified via normal phase chromatography (O ⁇ 10% MeOH in EtOAc) to afford the title compound (5-3) as a bone powder.
  • the reaction mixture was cooled and diluted with ethyl acetate (30 mL).
  • the reaction mixture was washed with saturated sodium bicarbonate (2 x 10 mL), water (2 x 10 mL) and brine (1 x 10 mL), dried over magnesium sulfate and concentrated.
  • the residue was purified via normal phase chromatography (0 to 40% methanol in EtOAc, silica) followed by reverse phase chromatography (5 to 65% acetonitrile in water, 0.1% TFA buffer) to afford the desired product (6 ⁇ 4) as a white solid after free-basing and concentration.
  • Lithium aluminum hydride in diethyl ether (17.3 ml, 1.0 M, 17.3 mmol, 3.0 eq) was carefully added over twenty minutes to a solution of 3-cyclopropyl-4-methoxybenzonitrile (8-2, 1.Og, 5.77 mmol, 1.0 eq) in tetrahydrofuran (28.9 ml) at 0°C under nitrogen atmosphere.
  • the resulting dark orange mixture was allowed to stir at 0°C for 30 min, then carefully quenched in the following order: water (1.OmI), 15% NaOH aqueous (1.0 ml) and water (3.0 ml).
  • the resulting emulsion was stirred at room temperature for 30 min.
  • 6-Bromo-5-methoxvpyridine-2-carbonitrile (9-2)
  • 2-bromo-6-iodo-3-methoxypryidine (9-1 or 1 1-3. 2.1 g, 6.69 mmol, 1.0 equiv) in dichloromethane (17 mL) was added isopropylmagnesium chloride (2.0 M, 4.35 mL, 8.70 mmol, 1.3 equiv) slowly at 0 °C and the reaction mixture was stirred for 45 minutes.
  • 6-bromo-5-methoxypyridine-2-carbonitrile 9-2, 200 mg, 0.94 mmol, 1.0 equiv
  • potassium trifluoro(vinyl)borate 314 mg, 2.35 mmol, 2.5 equiv
  • tricyclohexylphosphine 52.7 mg, 0.19 mmol, 0.2 equiv
  • palladium(II) acetate 21.1 mg, 0.09 mmol, 0.1 equiv
  • tripotassium phosphate 697 mg, 3.29 mmol, 3.5 equiv
  • 6-bromo-5-methoxypyridine-2-carbonitrile 9 ⁇ , 300 mg, 1.41 mmol, 1.0 equiv
  • potassium cyclopropyl(trifluoro)borate 521 mg, 3.52 mmol, 2.5 equiv
  • tricyclohexylphosphine 158 mg, 0.56 mmol, 0.4 equiv
  • palladium(II) acetate 63.2 mg, 0.28 mmol, 0.2 equiv
  • tripotassium phosphate 1046 mg, 4.93 mmol, 3.5 equiv
  • 6-bromo-5-methoxypyridine-2-carbonitrile 15-1 or 9-2, 200 mg, 0.94 mmol, 1.0 equiv
  • potassium trifluoro(isopropenyl)borate (Molander, Gary A., J. Am. Chem. Soc. Commun.
  • 6-Ethoxy-5-methoxypyridine-2-carbonitrile (16-3) To a solution of 2-ethoxy-6-iodo-3-methoxypyridine (16-2, 100 mg, 0.36 mmol, 1 equiv) in DMF (675 uL) was added copper cyanide (39 mg, 0.43, mmol, 1.2 equiv). The reaction mixture was heated in a microwave reactor at 150°C for 20 minutes, and then was partitioned between DCM (5OmL) and water (50 mL).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Epidemiology (AREA)
  • Cardiology (AREA)
  • Urology & Nephrology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pulmonology (AREA)
  • Diabetes (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Endocrinology (AREA)
  • Pain & Pain Management (AREA)
  • Hospice & Palliative Care (AREA)
  • Reproductive Health (AREA)
  • Rheumatology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Hematology (AREA)
  • Psychiatry (AREA)
  • Obesity (AREA)
  • Anesthesiology (AREA)
  • Psychology (AREA)
  • Child & Adolescent Psychology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Nutrition Science (AREA)
  • Otolaryngology (AREA)

Abstract

The present invention is directed to pyridyl carboxamide compounds which are antagonists of orexin receptors, and which are useful in the treatment or prevention of neurological and psychiatric disorders and diseases in which orexin receptors are involved. The invention is also directed to pharmaceutical compositions comprising these compounds and the use of these compounds and compositions in the prevention or treatment of such diseases in which orexin receptors are involved.

Description

TITLE OF THE INVENTION
PYRIDINE CARBOXAMIDE OREXIN RECEPTOR ANTAGONISTS
BACKGROUND OF THE INVENTION The orexins (hypocretins) comprise two neuropeptides produced in the hypothalamus: the orexin A (OX-A) (a 33 amino acid peptide) and the orexin B (OX-B) (a 28 amino acid peptide) (Sakurai T. et al., Cell, 1998, 92, 573-585). Orexins are found to stimulate food consumption in rats suggesting a physiological role for these peptides as mediators in the central feedback mechanism that regulates feeding behaviour (Sakurai T. et al., Cell, 1998, 92, 573-585). Orexins regulate states of sleep and wakefulness opening potentially novel therapeutic approaches for narcoleptic or insomniac patients (Chemelli R.M. et al., Cell, 1999, 98, 437-451). Orexins have also been indicated as playing a role in arousal, reward, learning and memory (Harris, et al., Trends Neurosci., 2006, 29 (10), 571-577). Two orexin receptors have been cloned and characterized in mammals. They belong to the super family of G-protein coupled receptors (Sakurai T. et al., Cell, 1998, 92, 573-585): the orexin- 1 receptor (OX or OXlR) is selective for OX-A and the orexin-2 receptor (0X2 or 0X2R) is capable to bind OX-A as well as OX-B. The physiological actions in which orexins are presumed to participate are thought to be expressed via one or both of OX 1 receptor and OX 2 receptor as the two subtypes of orexin receptors. Orexin receptors are found in the mammalian brain and may have numerous implications in pathologies such as depression; anxiety; addictions; obsessive compulsive disorder; affective neurosis; depressive neurosis; anxiety neurosis; dysthymic disorder; behaviour disorder; mood disorder; sexual dysfunction; psychosexual dysfunction; sex disorder; schizophrenia; manic depression; delirium; dementia; severe mental retardation and dyskinesias such as Huntington's disease and Tourette syndrome; eating disorders such as anorexia, bulimia, cachexia, dysregulated appetite control; obesity; addictive feeding behaviors; binge/purge feeding behaviors; cardiovascular diseases; diabetes; appetite, taste, eating or drinking disorders; emesis, vomiting, nausea; asthma; cancer; Parkinson's disease; Cushing's syndrome/disease; basophile adenoma; prolactinoma; hyperprolactinemia; hypophysis tumour/adenoma; hypothalamic diseases; inflammatory bowel disease; gastric diskinesia; gastric ulcers; Froehlich's syndrome; adrenohypophysis disease; hypophysis disease; adrenohypophysis hypofunction; adrenohypophysis hyperfunction; hypothalamic hypogonadism; Kallman's syndrome (anosmia, hyposmia); functional or psychogenic amenorrhea; hypopituitarism; hypothalamic hypothyroidism; hypothalamic- adrenal dysfunction; idiopathic hyperprolactinemia; hypothalamic disorders of growth hormone deficiency; idiopathic growth deficiency; dwarfism; gigantism; acromegaly; disturbed biological and circadian rhythms; sleep disturbances associated with diseases such as neurological disorders, neuropathic pain and restless leg syndrome; heart and lung diseases, acute and congestive heart failure; hypotension; hypertension; urinary retention; osteoporosis; angina pectoris; myocardinal infarction; ischemic or haemorrhagic stroke; subarachnoid haemorrhage; ulcers; allergies; benign prostatic hypertrophy; chronic renal failure; renal disease; impaired glucose tolerance; migraine; hyperalgesia; pain; enhanced or exaggerated sensitivity to pain such as hyperalgesia, causalgia, and allodynia; acute pain; burn pain; atypical facial pain; neuropathic pain; back pain; complex regional pain syndrome I and II; arthritic pain; sports injury pain; pain related to infection e.g. HIV, post-chemotherapy pain; post- stroke pain; post-operative pain; neuralgia; conditions associated with visceral pain such as irritable bowel syndrome, and angina; urinary bladder incontinence e.g. urge incontinence; tolerance to narcotics or withdrawal from narcotics; sleep disorders; sleep apnea; narcolepsy; insomnia; parasomnia; jet lag syndrome; and neurodegenerative disorders including nosological entities such as disinhibition-dementia-parkinsonism-amyotrophy complex; pallido-ponto-nigral degeneration; epilepsy; seizure disorders and other diseases related to general orexin system dysfunction. Certain orexin receptor antagonists are disclosed in PCT patent publications WO
99/09024, WO 99/58533, WO 00/47576, WO 00/47577, WO 00/47580, WO 01/68609, WO 01/85693, WO 01/96302, WO 2002/044172, WO 2002/051232, WO 2002/051838, WO 2002/089800, WO 2002/090355, WO 2003/002559, WO 2003/002561, WO 2003/032991, WO 2003/037847, WO 2003/041711, WO 2003/051368, WO 2003/051872, WO 2003/051873, WO 2004/004733, WO 2004/026866, WO 2004/033418, WO 2004/041807, WO 2004/041816, WO 2004/052876, WO 2004/083218, WO 2004/085403, WO 2004/096780, WO 2005/060959, WO 2005/075458, WO2005/1 18548, WO 2006/067224, WO 2006/1 10626, WO 2006/127550, WO 2007/019234, WO 2007/025069, WO 2007/061763, WO 2007/1 16374, WO 2007/122591, WO 2007/126934, WO 2007/126935, WO 2008/008517, WO 2008/008518, WO 2008/008551, WO 2008/020405, WO 2008//026149, WO 2008/038251.
SUMMARY OF THE INVENTION
The present invention is directed to pyridyl carboxamide compounds which are antagonists of orexin receptors, and which are useful in the treatment or prevention of neurological and psychiatric disorders and diseases in which orexin receptors are involved. The invention is also directed to pharmaceutical compositions comprising these compounds and the use of these compounds and compositions in the prevention or treatment of such diseases in which orexin receptors are involved.
DETAILED DESCRIPTION OF THE INVENTION
The present invention is directed to compounds of the formula I:
Figure imgf000004_0001
wherein:
A1 is selected from the group consisting of phenyl, naphthyl and heteroaryl;
A2 is selected from the group consisting of phenyl, naphthyl and heteroaryl;
A3 is selected from the group consisting of phenyl, naphthyl, C3-6cycloalkyl, and heterocycle;
R1 a, R1b and R1 c may be absent if the valency of A1 does not permit such substitution and are independently selected from the group consisting of:
(1) hydrogen,
(2) halogen,
(3) hydroxyl, (4) -(C=O)m-On-C1 -6alkyl, where m is 0 or 1 , n is 0 or 1 (wherein if m is 0 or n is 0, a bond is present) and where the alkyl is unsubstituted or substituted with one or more substituents selected from R13, (5) -(C=O)m-On-C3-6cycloalkyl, where the cycloalkyl is unsubstituted or substituted with one or more substituents selected from R13, (6) -(C=O)m-C2-4alkenyl, where the alkenyl is unsubstituted or substituted with one or more substituents selected from R13,
(7) -(C=O)m-C2-4alkynyl, where the alkynyl is unsubstituted or substituted with one or more substituents selected from R13,
(8) -(C=O)m-On-phenyl or -(C=O)m-On-napthyl, where the phenyl or naphthyl is unsubstituted or substituted with one or more substituents selected from R13,
(9) -(C=O)m-On-heterocycle, where the heterocycle is unsubstituted or substituted with one or more substituents selected from R13, (10) -(C=0)m-NR10R1 1, wherein R10 and R1 1 are independently selected from the group consisting of:
(a) hydrogen,
(b) C1 -6alkyl, which is unsubstituted or substituted with R13, (c) C3-6alkenyl, which is unsubstituted or substituted with R13,
(d) C3-6alkynyl, which is unsubstituted or substituted with R13,
(e) C3-6cycloalkyl which is unsubstituted or substituted with R13,
(f) phenyl, which is unsubstituted or substituted with R13, and
(g) heterocycle, which is unsubstituted or substituted with R13, (1 1) -S(0)2-NR10R1 l ,
(12) -S(O)q-R12, where q is 0, 1 or 2 and where R12 is selected from the definitions of R1O and R1 I,
(13) -CO2H,
(14) -CN, and (15) -Nθ2;
R2a, R2b and R2c may be absent if the valency of A2 does not permit such substitution and are independently selected from the group consisting of:
(1) hydrogen, (2) halogen,
(3) hydroxyl,
(4) -(C=O)m-On-C1 -6alkyl, where the alkyl is unsubstituted or substituted with one or more substituents selected from R13,
(5) -(C=O)m-On-C3-6cycloalkyl, where the cycloalkyl is unsubstituted or substituted with one or more substituents selected from R13,
(6) -(C=O)m-C2-4alkenyl, where the alkenyl is unsubstituted or substituted with one or more substituents selected from R13,
(7) -(C=O)m-C2-4alkynyl, where the alkynyl is unsubstituted or substituted with one or more substituents selected from R13, (8) -(C=O)m-On-phenyl or -(C=O)m-On-napthyl, where the phenyl or naphthyl is unsubstituted or substituted with one or more substituents selected from R13,
(9) -(C=O)m-On-heterocycle, where the heterocycle is unsubstituted or substituted with one or more substituents selected from R13,
(10) -(C=O)1n-NR10R1 1 , (1 1) -S(0)2-NR1θR1 l ,
(12) -S(O)q-R12,
(13) -CO2H, (14) -CN, and
(15) -NO2;
R3a, R3b and R3c may be absent if the valency of A^ does not permit such substitution and are independently selected from the group consisting of:
(1) hydrogen,
(2) halogen,
(3) hydroxyl,
(4) -(C=O)m-On-C1 -6alkyl, where the alkyl is unsubstituted or substituted with one or more substituents selected from R13,
(5) -(C=O)m-On-C3-6cycloalkyl, where the cycloalkyl is unsubstituted or substituted with one or more substituents selected from R13,
(6) -(C=O)m-C2-4alkenyl, where the alkenyl is unsubstituted or substituted with one or more substituents selected from R13, (7) -(C=O)m-C2-4alkynyl, where the alkynyl is unsubstituted or substituted with one or more substituents selected from R13,
(8) -(C=O)m-On-phenyl or -(C=O)m-On-napthyl, where the phenyl or naphthyl is unsubstituted or substituted with one or more substituents selected from R13,
(9) -(C=O)m-Orrheterocycle, where the heterocycle is unsubstituted or substituted with one or more substituents selected from R13,
(10) -(C=O)1n-NR10R1 1 ,
(1 1) -S(0)2-NR1θR1 l ,
(12) -S(O)q-R12,
(13) -CO2H, (14) -CN, and
(15) -NO2; with the proviso that if A3 is pyridyl, at least one of R3a, R3b and R3c is other than hydrogen;
R4 and R5 are independently selected from hydrogen and C1 -6alkyl, which is unsubstituted or substituted with one or more substituents selected from R13, or R4 and R5 may be joined together to form a C3-6cycloalkyl with the carbon atom to which they are attached, where the cycloalkyl is unsubstituted or substituted with one or more substituents selected from R13;
R6 is hydrogen, C1 -6alkyl or C3-6cycloalkyl, which is unsubstituted or substituted with one or more substituents selected from R13;
R13 is selected from the group consisting of: (1) halogen,
(2) hydroxyl,
(3) -(C=O)1n-On-C 1 -6alkyl, where the alkyl is unsubstituted or substituted with one or more substituents selected from R14, (4) -On-(C i-3)perfluoroalkyl,
(5) -(C=O)m-On-C3-6cycloalkyl, where the cycloalkyl is unsubstituted or substituted with one or more substituents selected from R 14,
(6) -(C=O)m-C2-4alkenyl, where the alkenyl is unsubstituted or substituted with one or more substituents selected from R 14, (7) -(C=O)m-C2-4alkynyl, where the alkynyl is unsubstituted or substituted with one or more substituents selected from R 14,
(8) -(C=O)m-On-phenyl or -(C=O)m-On-napthyl, where the phenyl or naphthyl is unsubstituted or substituted with one or more substituents selected from R14,
(9) -(C=O)m-On-heterocycle, where the heterocycle is unsubstituted or substituted with one or more substituents selected from R 14,
(10) -(C-O)1n-NR10R1 1 ,
(11) -S(0)2-NR1θR1 l,
(12) -S(0)q-R12,
(13) -CO2H, (14) -CN, and
(15) -NO2;
R14 is selected from the group consisting of:
(I) hydroxyl, (2) halogen,
(3) C1 -6alkyl,
(4) -Cs-όcycloalkyl,
(5) -O-C1 -6alkyl,
(6) -0(C=O)-C 1 -6alkyl, (7) -NH-C1 -6alkyl,
(8) phenyl,
(9) heterocycle,
(10) -CO2H, and
(I I) -CN; or a pharmaceutically acceptable salt thereof.
An embodiment of the present invention includes compounds of the formula Ia:
Figure imgf000008_0001
Ia wherein A1, A2, A3, R 1 a, R1 b, RI C, R2a, R2b, R2C, R3a, R3b, R3C and R4 are defined herein; or a pharmaceutically acceptable salt thereof.
An embodiment of the present invention includes compounds of the formula Ib:
Figure imgf000008_0002
Ib wherein A1, A2, A3, RIa, R1 b, R 1 c ; R2a, R2b, R2C, R3a, R3b and R3c are defined herein; or a pharmaceutically acceptable salt thereof. An embodiment of the present invention includes compounds of the formula Ic:
Figure imgf000009_0001
wherein A2, A3, R Ia, R1 b, R Ic, R2a, R2b, R2C, R3a, R3b and R3c are defined herein; or a pharmaceutically acceptable salt thereof.
An embodiment of the present invention includes compounds of the formula Ic':
Figure imgf000009_0002
wherein A2, A3, R Ia, R1 b, R2a ; R2b, R2C, R3a, R3b and R3c are defined herein; or a pharmaceutically acceptable salt thereof.
An embodiment of the present invention includes compounds of the formula Id:
Figure imgf000009_0003
Id wherein A2, A3, R 1 a, R1 b, R 1 c, R2a, R2b, R2C, R3a, R3b and R3c are defined herein; or a pharmaceutically acceptable salt thereof.
An embodiment of the present invention includes compounds of the formula Id':
Figure imgf000010_0001
wherein A2, A3, R 1 a, R1 b, R 1 c, R2a, R2b, R2C, R3a, R3b and R3c are defined herein; or a pharmaceutically acceptable salt thereof.
An embodiment of the present invention includes compounds of the formula Ie:
Figure imgf000010_0002
wherein A3, R 1 a, R1 b, RI C, R2a, R2b, R2C, R3a, R3b and R3c are defined herein; or a pharmaceutically acceptable salt thereof.
An embodiment of the present invention includes compounds of the formula If:
Figure imgf000011_0001
wherein A3, R 1 a, RIb3 RI C, R.2a, R2b, R2C, R3a, R3b and R3c are defined herein; or a pharmaceutically acceptable salt thereof.
An embodiment of the present invention includes compounds of the formula Ig:
Figure imgf000011_0002
wherein A2, R 1 a, R1 b, RI C, R2a ; R2b, R2C, R3a, R3b and R3c are defined herein; or a pharmaceutically acceptable salt thereof. An embodiment of the present invention includes compounds of the formula Ih:
Figure imgf000012_0001
wherein A1, A2, R Ia, R1 b, R Ic, R2a, R2b, R2C, R3a, R3b and R3c are defined herein; or a pharmaceutically acceptable salt thereof.
An embodiment of the present invention includes compounds of the formula Ii:
Figure imgf000012_0002
wherein A1, A2, R 1 a, R1 b ; RI C, R2a, R2b, R2C, R3a, R3b and R3c are defined herein; or a pharmaceutically acceptable salt thereof. An embodiment of the present invention includes compounds of the formula Ij:
Figure imgf000013_0001
wherein A1, A2, R 1 a, R1 b, R 1 c, R.2a, R2b, R2C, R3a and R3b are defined herein; or a pharmaceutically acceptable salt thereof.
An embodiment of the present invention includes compounds of the formula Ik:
Figure imgf000013_0002
wherein A1, A2, R 1 a, R1 b, R 1 c, R2a, R2b, R2C, R3a and R3b are defined herein; or a pharmaceutically acceptable salt thereof. An embodiment of the present invention includes compounds of the formula II:
Figure imgf000014_0001
wherein A1, A2, R 1 a, R1 b, R 1 c, R2a, R2b and R2c are defined herein; or a pharmaceutically acceptable salt thereof.
An embodiment of the present invention includes compounds of the formula Im:
Figure imgf000014_0002
wherein A1, A2, R 1 a, R1 b ; R 1 c, R2a, R2b and R2c are defined herein; or a pharmaceutically acceptable salt thereof. An embodiment of the present invention includes compounds wherein A1 is phenyl. An embodiment of the present invention includes compounds wherein A1 is naphthyl. An embodiment of the present invention includes compounds wherein A1 is heteroaryl. An embodiment of the present invention includes compounds wherein A1 is pyridyl.
An embodiment of the present invention includes compounds wherein A2 is phenyl. An embodiment of the present invention includes compounds wherein A2 is naphthyl. An embodiment of the present invention includes compounds wherein A2 is heteroaryl. An embodiment of the present invention includes compounds wherein A2 is quinoxalinyl. An embodiment of the present invention includes compounds wherein A2 is pyridyl. An embodiment of the present invention includes compounds wherein A2 is pyrazinly. An embodiment of the present invention includes compounds wherein A 2 is quinolinyl. An embodiment of the present invention includes compounds wherein A2 is indolyl. An embodiment of the present invention includes compounds wherein A2 is dihydroindolyl. An embodiment of the present invention includes compounds wherein A2 is benzimidazolyl. An embodiment of the present invention includes compounds wherein A3 is pyrazolyl. An embodiment of the present invention includes compounds wherein A3 is pyrazolyl which is substituted with methyl. An embodiment of the present invention includes compounds wherein A3 is pyridyl, which is substituted with at least one substituent other than hydrogen. An embodiment of the present invention includes compounds wherein A3 is pyridyl, which is substituted with fluroro or chloro. An embodiment of the present invention includes compounds wherein A3 is phenyl. An embodiment of the present invention includes compounds wherein A3 is phenyl, which is substituted with at least one substituent other than hydrogen. An embodiment of the present invention includes compounds wherein A3 is phenyl, which is substituted with hydroxyl, methylaminocarbonyl or dimethylaminomethyl. An embodiment of the present invention includes compounds wherein A3 is cyclopentyl. An embodiment of the present invention includes compounds wherein A3 is morpholinyl. An embodiment of the present invention includes compounds wherein A3 is pyrrolidinyl. An embodiment of the present invention includes compounds wherein A3 is azetidinyl.
An embodiment of the present invention includes compounds wherein R1 a, R1 b and R1 c are independently selected from the group consisting of:
(1) hydrogen,
(2) halogen,
(3) hydroxyl,
(4) C1 -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl, phenyl or napthyl,
(5) -O-C1 -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl or phenyl,
(6) heteroaryl, wherein heteroaryl is selected from pyrrolyl, imidazolyl, indolyl, pyridyl, and pyrimidinyl, which is unsubstituted or substituted with halogen, hydroxyl, C1 -6alkyl, -O-C1 -6alkyl or-NO2,
(7) phenyl, which is unsubstituted or substituted with halogen, hydroxyl, C1 -6alkyl, -O-C1 -6alkyl or-NO2,
(8) -O-phenyl, which is unsubstituted or substituted with halogen, hydroxyl, C1 -6alkyl, -O-C1 -6alkyl or-NO2, and (9) -NH-C1 -6alkyl, or -N(C I -6alkyl)(C1 -6alkyl), which is unsubstituted or substituted with halogen, hydroxyl, C1 -6alkyl, -O-C1 -6alkyl or-NO2- An embodiment of the present invention includes compounds wherein R 1 a, R1 b and R1 c are independently selected from the group consisting of:
(1) hydrogen,
(2) halogen, (3) hydroxy 1,
(4) C1 -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl or phenyl or napthyl, and
(5) -O-C1 -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl or phenyl. An embodiment of the present invention includes compounds wherein R 1 a, R1 b and R1 c are independently selected from the group consisting of:
(1) hydrogen,
(2) halogen, and
(3) C1 -6alkyl. An embodiment of the present invention includes compounds wherein R 1 a, R1 b and R1 c are independently selected from the group consisting of:
(1) hydrogen,
(2) chloro,
(3) fluroro, and (4) methyl.
An embodiment of the present invention includes compounds wherein A1 is phenyl and R 1 a, R1 b and R1 c are independently selected from the group consisting of:
(1) hydrogen,
(2) chloro, (3) fluroro, and
(4) methyl.
An embodiment of the present invention includes compounds wherein R2a, R2b and R2c are independently selected from the group consisting of:
(1) hydrogen, (2) halogen,
(3) hydroxyl,
(4) C1 -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl or phenyl or napthyl,
(5) -O-C1 -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl or phenyl, (6) heteroaryl, wherein heteroaryl is selected from pyrrolyl, imidazolyl, indolyl, pyridyl, and pyrimidinyl, which is unsubstituted or substituted with halogen, hydroxyl, C1 -6alkyl, -O-C1 -6alkyl or-NO2,
(7) phenyl, which is unsubstituted or substituted with halogen, hydroxyl, C1 -6alkyl, -O-C1 -6alkyl or-NO2,
(8) -O-phenyl, which is unsubstituted or substituted with halogen, hydroxyl, C1 -6alkyl, -O-C1 -6alkyl or-NO2, and
(9) -NH-C 1 -6alkyl, or -N(C1 -6alkyl)(C 1 -6alkyl), which is unsubstituted or substituted with halogen, hydroxyl, C1 -6alkyl, -O-C1 -6alkyl or-NO2- An embodiment of the present invention includes compounds wherein R2a ; R2b and R2c are independently selected from the group consisting of:
(1) hydrogen,
(2) halogen,
(3) hydroxyl, (4) C l -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl or phenyl,
(5) -O-C1 -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl or phenyl, and
(6) -NH-C 1 -6alkyl, or -N(C l -6alkyl)(C l -6alkyl), which is unsubstituted or substituted with halogen. An embodiment of the present invention includes compounds wherein R2a, R2b and R2c are independently selected from the group consisting of:
(1) hydrogen,
(2) halogen,
(3) C1 -6alkyl, which is unsubstituted or substituted with halogen, (4) -O-C1 -6alkyl, which is unsubstituted or substituted with halogen, and
(5) -NH-C1 -6alkyl, or -N(C 1 -6alkyl)(C I -6alkyl), which is unsubstituted or substituted with halogen.
An embodiment of the present invention includes compounds wherein R2a ; R2b and R2c are independently selected from the group consisting of: (1) hydrogen,
(2) chloro,
(3) fluoro,
(4) bromo,
(5) methoxy, (6) t-butoxy,
(7) difluoromethyl, and
(8) trifiuoromethyl, (9) -N(CH3).
An embodiment of the present invention includes compounds wherein A2 is phenyl and R2a, R2b and R2c are independently selected from the group consisting of:
(1) hydrogen, (2) chloro,
(3) fluoro,
(4) bromo,
(5) methoxy,
(6) t-butoxy, (7) difluoromethyl, and
(8) trifluoromethyl,
(9) -N(CH3).
An embodiment of the present invention includes compounds wherein R3a, R3b and R3c are independently selected from the group consisting of: (1) hydrogen,
(2) halogen,
(3) hydroxyl,
(4) C1 -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl, phenyl or napthyl, (5) -O-C1 -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl or phenyl,
(6) heteroaryl, wherein heteroaryl is selected from pyrrolyl, imidazolyl, indolyl, pyridyl, and pyrimidinyl, which is unsubstituted or substituted with halogen, hydroxyl, Ci-όalkyl, -O-C1 -6alkyl or-NO2, (7) phenyl, which is unsubstituted or substituted with halogen, hydroxyl, C1 -6alkyl,
-O-C1 -6alkyl or-NO2,
(8) -O-phenyl, which is unsubstituted or substituted with halogen, hydroxyl, C1 -6alkyl, -O-C1 -6alkyl or-NO2, and
(9) -NH-C 1 -6alkyl, or -N(C I -6alkyl)(C 1 -6alkyl), which is unsubstituted or substituted with halogen, hydroxyl, C I -6alkyl, -O-C 1 -6alkyl or-NO2; with the proviso that if A^ is pyridyl, at least one of R3a, R3b and R3c is other than hydrogen.
An embodiment of the present invention includes compounds wherein R3a ; R.3b and R3c are independently selected from the group consisting of:
(1) hydrogen, (2) halogen,
(3) hydroxyl, (4) C1 -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl or phenyl or napthyl, and
(5) -O-C1 -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl or phenyl; with the proviso that if A3 is pyridyl, at least one of R3a, R.3b and R3C is other than hydrogen.
An embodiment of the present invention includes compounds wherein R?a, R3b and R3c are independently selected from the group consisting of:
(1) hydrogen,
(2) halogen, and (3) C1 -6alkyl; with the proviso that if A3 is pyridyl, at least one of R3a, R.3b and R3c is other than hydrogen.
An embodiment of the present invention includes compounds wherein R3a ; R3b and R3c are independently selected from the group consisting of:
(1) hydrogen, (2) chloro,
(3) fluroro, and
(4) methyl; with the proviso that if A3 is pyridyl, at least one of R3a, R3b and R3c is other than hydrogen.
An embodiment of the present invention includes compounds wherein A3 is pyridyl and wherein R3a js halogen, R3b is hydrogen and R3c is hydrogen. An embodiment of the present invention includes compounds wherein A3 is pyridyl and wherein R3a is chloro or fluoro, R3b is hydrogen and R3c is hydrogen.
An embodiment of the present invention includes compounds wherein A3 is pyrazolyl and wherein R3a is C1 -6alkyl, R3b is hydrogen and R3c is hydrogen. An embodiment of the present invention includes compounds wherein A3 is pyrazolyl and wherein R3a is methyl, R3b is hydrogen and R3c is hydrogen.
An embodiment of the present invention includes compounds wherein R4 is hydrogen or C1 -6alkyl. An embodiment of the present invention includes compounds wherein
R4 is hydrogen or methyl. An embodiment of the present invention includes compounds wherein
R4 is hydrogen. An embodiment of the present invention includes compounds wherein R^ is hydrogen or C1 -6alkyl. An embodiment of the present invention includes compounds wherein
R5 is hydrogen or methyl. An embodiment of the present invention includes compounds wherein
R5 is hydrogen.
An embodiment of the present invention includes compounds wherein R6 is hydrogen, C1 -6alkyl or C3-6cycloalkyl. An embodiment of the present invention includes compounds wherein R6 is C1 -6alkyl. An embodiment of the present invention includes compounds wherein R6 is C3-6cycloalkyl. An embodiment of the present invention includes compounds wherein R6 is hydrogen, methyl or ethyl. An embodiment of the present invention includes compounds wherein R6 is hydrogen.
Specific embodiments of the present invention include a compound which is selected from the group consisting of the subject compounds of the Examples herein or a pharmaceutically acceptable salt thereof.
The compounds of the present invention may contain one or more asymmetric centers and can thus occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. Additional asymmetric centers may be present depending upon the nature of the various substituents on the molecule. Each such asymmetric center will independently produce two optical isomers and it is intended that all of the possible optical isomers and diastereomers in mixtures and as pure or partially purified compounds are included within the ambit of this invention. The present invention is meant to comprehend all such isomeric forms of these compounds. Formula I shows the structure of the class of compounds without specific stereochemistry. The independent syntheses of these diastereomers or their chromatographic separations may be achieved as known in the art by appropriate modification of the methodology disclosed herein. Their absolute stereochemistry may be determined by the x-ray crystallography of crystalline products or crystalline intermediates which are derivatized, if necessary, with a reagent containing an asymmetric center of known absolute configuration. If desired, racemic mixtures of the compounds may be separated so that the individual enantiomers are isolated. The separation can be carried out by methods well known in the art, such as the coupling of a racemic mixture of compounds to an enantiomerically pure compound to form a diastereomeric mixture, followed by separation of the individual diastereomers by standard methods, such as fractional crystallization or chromatography. The coupling reaction is often the formation of salts using an enantiomerically pure acid or base. The diasteromeric derivatives may then be converted to the pure enantiomers by cleavage of the added chiral residue. The racemic mixture of the compounds can also be separated directly by chromatographic methods utilizing chiral stationary phases, which methods are well known in the art. A1ternatively, any enantiomer of a compound may be obtained by stereoselective synthesis using optically pure starting materials or reagents of known configuration by methods well known in the art.
As appreciated by those of skill in the art, halogen or halo as used herein are intended to include fluoro, chloro, bromo and iodo. Similarly, C1 -6, as in C1 -6alkyl is defined to identify the group as having 1, 2, 3, 4, 5 or 6 carbons in a linear or branched arrangement, such that Ci-8alkyl specifically includes methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert- butyl, pentyl, and hexyl. A group which is designated as being independently substituted with substituents may be independently substituted with multiple numbers of such substituents. The term "heterocycle" as used herein includes both unsaturated and saturated heterocyclic moieties, wherein the unsaturated heterocyclic moieties (i.e. "heteroaryl") include benzoimidazolyl, benzimidazolonyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzothiazolyl, benzotriazolyl, benzothiophenyl, benzoxazepin, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, imidazolyl, indolinyl, indolyl, dihydroindolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthpyridinyl, oxadiazolyl, oxazolyl, oxazoline, isoxazoline, oxetanyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridopyridinyl, pyridazinyl, pyridyl, pyrimidyl, pyrrolyl, quinazolinyl, quinolyl, quinoxalinyl, tetrahydroquinoxalinyl, tetrazolyl, tetrazolopyridyl, thiadiazolyl, thiazolyl, thienyl, triazolyl, and N-oxides thereof, and wherein the saturated heterocyclic moieties include azetidinyl, 1 ,4-dioxanyl, hexahydroazepinyl, piperazinyl, piperidinyl, pyridin-2-onyl, pyrrolidinyl, morpholinyl, tetrahydrofuranyl, thiomorpholinyl, and tetrahydrothienyl, and N-oxides thereof.
The term "pharmaceutically acceptable salts" refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids including inorganic or organic bases and inorganic or organic acids. Salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc, and the like. Particular embodiments include the ammonium, calcium, magnesium, potassium, and sodium salts. Salts in the solid form may exist in more than one crystal structure, and may also be in the form of hydrates. Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N'-dibenzylethylene-diamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethyl-morpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like.
When the compound of the present invention is basic, salts may be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids. Such acids include acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p- toluenesulfonic acid, and the like. Particular embodiments include the citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, fumaric, and tartaric acids. It will be understood that, as used herein, references to the compounds of Formula I are meant to also include the pharmaceutically acceptable salts.
Exemplifying the invention is the use of the compounds disclosed in the Examples and herein. Specific compounds within the present invention include a compound which selected from the group consisting of the compounds disclosed in the following Examples and pharmaceutically acceptable salts thereof and individual enantiomers or diastereomers thereof.
The subject compounds are useful in a method of antagonizing orexin receptor activity in a patient such as a mammal in need of such inhibition comprising the administration of an effective amount of the compound. The present invention is directed to the use of the compounds disclosed herein as antagonists of orexin receptor activity. In addition to primates, especially humans, a variety of other mammals can be treated according to the method of the present invention. The present invention is directed to a compound of the present invention or a pharmaceutically acceptable salt thereof for use in medicine. The present invention is further directed to a use of a compound of the present invention or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for antagonizing orexin receptor activity or treating the disorders and diseases noted herein in humans and animals.
The subject treated in the present methods is generally a mammal, such as a human being, male or female. The term "therapeutically effective amount" means the amount of the subject compound that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician. It is recognized that one skilled in the art may affect the neurological and psychiatric disorders by treating a patient presently afflicted with the disorders or by prophylactically treating a patient afflicted with the disorders with an effective amount of the compound of the present invention. As used herein, the terms "treatment" and "treating" refer to all processes wherein there may be a slowing, interrupting, arresting, controlling, or stopping of the progression of the neurological and psychiatric disorders described herein, but does not necessarily indicate a total elimination of all disorder symptoms, as well as the prophylactic therapy of the mentioned conditions, particularly in a patient who is predisposed to such disease or disorder. The terms "administration of and or "administering a" compound should be understood to mean providing a compound of the invention or a prodrug of a compound of the invention to the individual in need thereof.
The term "composition" as used herein is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. Such term in relation to pharmaceutical composition, is intended to encompass a product comprising the active ingredient(s), and the inert ingredient(s) that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. Accordingly, the pharmaceutical compositions of the present invention encompass any composition made by admixing a compound of the present invention and a pharmaceutically acceptable carrier. By "pharmaceutically acceptable" it is meant the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. The utility of the compounds in accordance with the present invention as orexin receptor OXlR and/or 0X2R antagonists may be readily determined without undue experimentation by methodology well known in the art, including the "FLIPR Ca2+ Flux Assay" (Okumura et al., Biochem. Biophys. Res. Comm. 280:976-981, 2001). In a typical experiment the OXl and 0X2 receptor antagonistic activity of the compounds of the present invention was determined in accordance with the following experimental method. For intracellular calcium measurements, Chinese hamster ovary (CHO) cells expressing the rat orexin- 1 receptor or the human orexin-2 receptor, are grown in Iscove's modified DMEM containing 2 mM L-glutamine, 0.5 g/ml G418, 1% hypoxanthine-thymidine supplement, 100 U/ml penicillin, 100 ug/ml streptomycin and 10 % heat-inactivated fetal calf serum (FCS). The cells are seeded at 20,000 cells / well into Becton-Dickinson black 384-well clear bottom sterile plates coated with poly-D- lysine. All reagents were from GIBCO-Invitrogen Corp. The seeded plates are incubated overnight at 37°C and 5% CO2. Ala-6,12 human orexin-A as the agonist is prepared as a 1 mM stock solution in 1% bovine serum albumin (BSA) and diluted in assay buffer (HBSS containing 20 mM HEPES, 0.1% BSA and 2.5mM probenecid, pH7.4) for use in the assay at a final concentration of 7OpM. Test compounds are prepared as 10 mM stock solution in DMSO, then diluted in 384-well plates, first in DMSO, then assay buffer. On the day of the assay, cells are washed 3 times with 100 ul assay buffer and then incubated for 60 min (37° C, 5% CO2) in 60 ul assay buffer containing 1 uM Fluo-4AM ester, 0.02 % pluronic acid, and 1 % BSA. The dye loading solution is then aspirated and cells are washed 3 times with 100 ul assay buffer. 30 ul of that same buffer is left in each well. Within the Fluorescent Imaging Plate Reader (FLIPR,
Molecular Devices), test compounds are added to the plate in a volume of 25 ul, incubated for 5 min and finally 25 ul of agonist is added. Fluorescence is measured for each well at 1 second intervals for 5 minutes and the height of each fluorescence peak is compared to the height of the fluorescence peak induced by 70 pM Ala-6,12 orexin-A with buffer in place of antagonist. For each antagonist, IC50 value (the concentration of compound needed to inhibit 50 % of the agonist response) is determined. A1ternatively, compound potency can be assessed by a radioligand binding assay (described in Bergman et. al. Bioorg. Med. Chem. Lett. 2008, 18, 1425 - 1430) in which the inhibition constant (Kj) is determined in membranes prepared from CHO cells expressing either the OXl or OX2 receptor. The intrinsic orexin receptor antagonist activity of a compound which may be used in the present invention may be determined by these assays. In particular, the compounds of the following examples had activity in antagonizing the rat orexin-1 receptor and/or the human orexin-2 receptor in the aforementioned assays, generally with an IC50 of less than about 50 μM. Many of compounds within the present invention had activity in antagonizing the rat orexin-1 receptor and/or the human orexin-2 receptor in the aforementioned assays with an IC50 of less than about 100 nM. Compounds of the present invention also have activity in the radioligand binding assay, generally with a Ki < 100 nM against the orexin-1 and/or the orexin-2 receptor. Additional data is provided in the following Examples. Such a result is indicative of the intrinsic activity of the compounds in use as antagonists of orexin-1 receptor and/or the orexin-2 receptor. The present invention also includes compounds within the generic scope of the invention which possess activity as agonists of the orexin-1 receptor and/or the orexin-2 receptor. With respect to other pyridyl compounds, the present compounds exhibit unexpected properties, such as with respect to increased oral bioavailability, metabolic stability, decreased inhibition of metabolic enzymes (such as decreased cytochrome P450 3A4 (CYP3A4) inhibition), decreased inhibition of transporters (such as decreased p-glycoprotein/PGP inhibition) and/or selectivity with respect to other receptors, including the human orexin-2 receptor.
The orexin receptors have been implicated in a wide range of biological functions. This has suggested a potential role for these receptors in a variety of disease processes in humans or other species. The compounds of the present invention have utility in treating, preventing, ameliorating, controlling or reducing the risk of a variety of neurological and psychiatric disorders associated with orexin receptors, including one or more of the following conditions or diseases: sleep disorders, sleep disturbances, including enhancing sleep quality, improving sleep quality, increasing sleep efficiency, augmenting sleep maintenance; increasing the value which is calculated from the time that a subject sleeps divided by the time that a subject is attempting to sleep; improving sleep initiation; decreasing sleep latency or onset (the time it takes to fall asleep); decreasing difficulties in falling asleep; increasing sleep continuity; decreasing the number of awakenings during sleep; decreasing intermittent wakings during sleep; decreasing nocturnal arousals; decreasing the time spent awake following the initial onset of sleep; increasing the total amount of sleep; reducing the fragmentation of sleep; altering the timing, frequency or duration of REM sleep bouts; altering the timing, frequency or duration of slow wave (i.e. stages 3 or 4) sleep bouts; increasing the amount and percentage of stage 2 sleep; promoting slow wave sleep; enhancing EEG-delta activity during sleep; decreasing nocturnal arousals, especially early morning awakenings; increasing daytime alertness; reducing daytime drowsiness; treating or reducing excessive daytime sleepiness; increasing satisfaction with the intensity of sleep; increasing sleep maintenance; idiopathic insomnia; sleep problems; insomnia, hypersomnia, idiopathic hypersomnia, repeatability hypersomnia, intrinsic hypersomnia, narcolepsy, interrupted sleep, sleep apnea, wakefulness, nocturnal myoclonus, REM sleep interruptions, jet-lag, shift workers' sleep disturbances, dyssomnias, night terror, insomnias associated with depression, emotional/mood disorders, Alzheimer's disease or cognitive impairment, as well as sleep walking and enuresis, and sleep disorders which accompany aging; Alzheimer's sundowning; conditions associated with circadian rhythmicity as well as mental and physical disorders associated with travel across time zones and with rotating shift-work schedules, conditions due to drugs which cause reductions in REM sleep as a side effect; fibromyalgia; syndromes which are manifested by non-restorative sleep and muscle pain or sleep apnea which is associated with respiratory disturbances during sleep; conditions which result from a diminished quality of sleep; increasing learning; augmenting memory; increasing retention of memory; eating disorders associated with excessive food intake and complications associated therewith, compulsive eating disorders, obesity (due to any cause, whether genetic or environmental), obesity-related disorders including overeating and bulimia nervosa, hypertension, diabetes, elevated plasma insulin concentrations and insulin resistance, dyslipidemias, hyperlipidemia, endometrial, breast, prostate and colon cancer, osteoarthritis, obstructive sleep apnea, cholelithiasis, gallstones, heart disease, abnormal heart rhythms and arrythmias, myocardial infarction, congestive heart failure, coronary heart disease, sudden death, stroke, polycystic ovary disease, craniopharyngioma, the Prader-Willi Syndrome, Frohlich's syndrome, GH-deficient subjects, normal variant short stature, Turner's syndrome, and other pathological conditions showing reduced metabolic activity or a decrease in resting energy expenditure as a percentage of total fat-free mass, e.g, children with acute lymphoblastic leukemia, metabolic syndrome, also known as syndrome X, insulin resistance syndrome, reproductive hormone abnormalities, sexual and reproductive dysfunction, such as impaired fertility, infertility, hypogonadism in males and hirsutism in females, fetal defects associated with maternal obesity, gastrointestinal motility disorders, intestinal motility dyskinesias, obesity- related gastro-esophageal reflux, hypothalmic diseases, hypophysis diseases, respiratory disorders, such as obesity-hypoventilation syndrome (Pickwickian syndrome), breathlessness, cardiovascular disorders, inflammation, such as systemic inflammation of the vasculature, arteriosclerosis, hypercholesterolemia, hyperuricaemia, lower back pain, gallbladder disease, gout, kidney cancer, increased anesthetic risk, reducing the risk of secondary outcomes of obesity, such as reducing the risk of left ventricular hypertrophy; diseases or disorders where abnormal oscillatory activity occurs in the brain, including depression, migraine, neuropathic pain, Parkinson's disease, psychosis and schizophrenia, as well as diseases or disorders where there is abnormal coupling of activity, particularly through the thalamus; enhancing cognitive function, including cognitive dysfunctions that comprise deficits in all types of attention, learning and memory functions occuring transiently or chronically in the normal, healthy, young, adult or aging population, and also occurring transiently or chronically in psychiatric, neurologic, cardiovascular and immune disorders; enhancing memory; increasing memory retention; increasing immune response; increasing immune function; hot flashes; night sweats; extending life span; schizophrenia; muscle-related disorders that are controlled by the excitation/relaxation rhythms imposed by the neural system such as cardiac rhythm and other disorders of the cardiovascular system; conditions related to proliferation of cells such as vasodilation or vasorestriction and blood pressure; cancer; cardiac arrhythmia; hypertension; congestive heart failure; conditions of the genital/urinary system; disorders of sexual function and fertility; adequacy of renal function; responsivity to anesthetics; mood disorders, such as depression or more particularly depressive disorders, for example, single episodic or recurrent major depressive disorders and dysthymic disorders, or bipolar disorders, for example, bipolar I disorder, bipolar II disorder and cyclothymic disorder, mood disorders due to a general medical condition, and substance-induced mood disorders; anxiety disorders including acute stress disorder, agoraphobia, generalized anxiety disorder, obsessive-compulsive disorder, panic attack, panic disorder, post-traumatic stress disorder, separation anxiety disorder, social phobia, specific phobia, substance-induced anxiety disorder and anxiety due to a general medical condition; acute neurological and psychiatric disorders such as cerebral deficits subsequent to cardiac bypass surgery and grafting, stroke, ischemic stroke, cerebral ischemia, spinal cord trauma, head trauma, perinatal hypoxia, cardiac arrest, hypoglycemic neuronal damage; Huntington's Chorea; amyotrophic lateral sclerosis; multiple sclerosis; ocular damage; retinopathy; cognitive disorders; idiopathic and drug-induced Parkinson's disease; muscular spasms and disorders associated with muscular spasticity including tremors, epilepsy, convulsions, seizure disorders, absence seisures, complex partial and generalized seizures; Lennox-Gastaut syndrome; cognitive disorders including dementia (associated with Alzheimer's disease, ischemia, trauma, vascular problems or stroke, HIV disease, Parkinson's disease, Huntington's disease, Pick's disease, Creutzfeldt- Jacob disease, perinatal hypoxia, other general medical conditions or substance abuse); delirium, amnestic disorders or age related cognitive decline; schizophrenia or psychosis including schizophrenia (paranoid, disorganized, catatonic or undifferentiated), schizophreniform disorder, schizoaffective disorder, delusional disorder, brief psychotic disorder, shared psychotic disorder, psychotic disorder due to a general medical condition and substance-induced psychotic disorder; dissociateive disorders including multiple personality syndromes and psychogenic amnesias; substance-related disorders, substance use, substance abuse, substance seeking, substance reinstatement, all types of psychological and physical addictions and addictive behaviors, reward- related behaviors (including substance-induced delirium, persisting dementia, persisting amnestic disorder, psychotic disorder or anxiety disorder; tolerance, addictive feeding, dependence, withdrawal or relapse from substances including alcohol, amphetamines, cannabis, cocaine, hallucinogens, inhalants, morphine, nicotine, opioids, phencyclidine, sedatives, hypnotics or anxiolytics); movement disorders, including akinesias and akinetic-rigid syndromes (including Parkinson's disease, drug-induced parkinsonism, postencephalitic parkinsonism, progressive supranuclear palsy, multiple system atrophy, corticobasal degeneration, parkinsonism-ALS dementia complex and basal ganglia calcification), chronic fatigue syndrome, fatigue, including Parkinson's fatigue, multiple sclerosis fatigue, fatigue caused by a sleep disorder or a circadian rhythm disorder, medication-induced parkinsonism (such as neuroleptic-induced parkinsonism, neuroleptic malignant syndrome, neuroleptic-induced acute dystonia, neuroleptic-induced acute akathisia, neuroleptic-induced tardive dyskinesia and medication-induced postural tremor), Gilles de Ia Tourette's syndrome, epilepsy, and dyskinesias [including tremor (such as rest tremor, essential tremor, postural tremor and intention tremor), chorea (such as Sydenham's chorea, Huntington's disease, benign hereditary chorea, neuroacanthocytosis, symptomatic chorea, drug- induced chorea and hemiballism), myoclonus (including generalised myoclonus and focal myoclonus), tics (including simple tics, complex tics and symptomatic tics), restless leg syndrome and dystonia (including generalised dystonia such as iodiopathic dystonia, drug- induced dystonia, symptomatic dystonia and paroxymal dystonia, and focal dystonia such as blepharospasm, oromandibular dystonia, spasmodic dysphonia, spasmodic torticollis, axial dystonia, dystonic writer's cramp and hemiplegic dystonia); attention deficit/hyperactivity disorder (ADHD); conduct disorder; migraine (including migraine headache); headache; hyperalgesia; pain; enhanced or exaggerated sensitivity to pain such as hyperalgesia, causalgia, and allodynia; acute pain; burn pain; atypical facial pain; neuropathic pain; back pain; complex regional pain syndrome I and II; arthritic pain; sports injury pain; pain related to infection e.g. HIV, post-chemotherapy pain; post-stroke pain; post-operative pain; neuralgia; emesis, nausea, vomiting; gastric dyskinesia; gastric ulcers; Kallman's syndrome (anosmia); conditions associated with visceral pain such as irritable bowel syndrome, and angina; eating disorders; urinary incontinence; substance tolerance, substance withdrawal (including, substances such as opiates, nicotine, tobacco products, alcohol, benzodiazepines, cocaine, sedatives, hypnotics, etc.); psychosis; schizophrenia; anxiety (including generalized anxiety disorder, panic disorder, and obsessive compulsive disorder); mood disorders (including depression, mania, bipolar disorders); trigeminal neuralgia; hearing loss; tinnitus; neuronal damage including ocular damage; retinopathy; macular degeneration of the eye; emesis; brain edema; pain, including acute and chronic pain states, severe pain, intractable pain, inflammatory pain, neuropathic pain, post- traumatic pain, bone and joint pain (osteoarthritis), repetitive motion pain, dental pain, cancer pain, myofascial pain (muscular injury, fibromyalgia), perioperative pain (general surgery, gynecological), chronic pain, neuropathic pain, post-traumatic pain, trigeminal neuralgia, migraine and migraine headache.
Thus, in specific embodiments the present invention provides methods for: enhancing the quality of sleep; augmenting sleep maintenance; increasing REM sleep; increasing stage 2 sleep; decreasing fragmentation of sleep patterns; treating insomnia and all types of sleep disorders; treating or controlling sleep disturbances associated with diseases such as neurological disorders including neuropathic pain and restless leg syndrome; treating or controlling addiction disorders; treating or controlling psychoactive substance use and abuse; enhancing cognition; increasing memory retention; treating or controlling obesity; treating or controlling diabetes and appetite, taste, eating, or drinking disorders; treating or controlling hypothalamic diseases; treating or controlling depression; treating, controlling, ameliorating or reducing the risk of epilepsy, including absence epilepsy; treating or controlling pain, including neuropathic pain; treating or controlling Parkinson's disease; treating or controlling psychosis; treating or controlling dysthymic, mood, psychotic and anxiety disorders; treating or controlling depression, including major depression and major dperession disorder; treating or controlling bipolar disorder; or treating, controlling, ameliorating or reducing the risk of schizophrenia, in a mammalian patient in need thereof which comprises administering to the patient a therapeutically effective amount of a compound of the present invention.
The subject compounds are further useful in a method for the prevention, treatment, control, amelioration, or reducation of risk of the diseases, disorders and conditions noted herein. The dosage of active ingredient in the compositions of this invention may be varied, however, it is necessary that the amount of the active ingredient be such that a suitable dosage form is obtained. The active ingredient may be administered to patients (animals and human) in need of such treatment in dosages that will provide optimal pharmaceutical efficacy. The selected dosage depends upon the desired therapeutic effect, on the route of administration, and on the duration of the treatment. The dose will vary from patient to patient depending upon the nature and severity of disease, the patient's weight, special diets then being followed by a patient, concurrent medication, and other factors which those skilled in the art will recognize. Generally, dosage levels of between 0.0001 to 10 mg/kg. of body weight daily are administered to the patient, e.g., humans and elderly humans, to obtain effective antagonism of orexin receptors. The dosage range will generally be about 0.5 mg to 1.0 g. per patient per day which may be administered in single or multiple doses. In one embodiment, the dosage range will be about 0.5 mg to 500 mg per patient per day; in another embodiment about 0.5 mg to 200 mg per patient per day; and in yet another embodiment about 5 mg to 50 mg per patient per day. Pharmaceutical compositions of the present invention may be provided in a solid dosage formulation such as comprising about 0.5 mg to 500 mg active ingredient, or comprising about 1 mg to 250 mg active ingredient. The pharmaceutical composition may be provided in a solid dosage formulation comprising about 1 mg, 5 mg, 10 mg, 25 mg, 30 mg, 50 mg, 80 mg, 100 mg, 200 mg or 250 mg active ingredient. For oral administration, the compositions may be provided in the form of tablets containing 1.0 to 1000 milligrams of the active ingredient, such as 1, 5, 10, 15, 20, 25, 50, 75, 100, 150, 200, 250, 300, 400, 500, 600, 750, 800, 900, and 1000 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. The compounds may be administered on a regimen of 1 to 4 times per day, such as once or twice per day. The compounds may be administered before bedtime. For example, the compounds may be administered about 1Hour prior to bedtime, about 30 minutes prior to bedtime or immediately before bedtime.
The compounds of the present invention may be used in combination with one or more other drugs in the treatment, prevention, control, amelioration, or reduction of risk of diseases or conditions for which compounds of the present invention or the other drugs may have utility, where the combination of the drugs together are safer or more effective than either drug alone. Such other drug(s) may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with a compound of the present invention. When a compound of the present invention is used contemporaneously with one or more other drugs, a pharmaceutical composition in unit dosage form containing such other drugs and the compound of the present invention is contemplated. However, the combination therapy may also includes therapies in which the compound of the present invention and one or more other drugs are administered on different overlapping schedules. It is also contemplated that when used in combination with one or more other active ingredients, the compounds of the present invention and the other active ingredients may be used in lower doses than when each is used singly. Accordingly, the pharmaceutical compositions of the present invention include those that contain one or more other active ingredients, in addition to a compound of the present invention. The above combinations include combinations of a compound of the present invention not only with one other active compound, but also with two or more other active compounds.
Likewise, compounds of the present invention may be used in combination with other drugs that are used in the prevention, treatment, control, amelioration, or reduction of risk of the diseases or conditions for which compounds of the present invention are useful. Such other drugs may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with a compound of the present invention. When a compound of the present invention is used contemporaneously with one or more other drugs, a pharmaceutical composition containing such other drugs in addition to the compound of the present invention is contemplated. Accordingly, the pharmaceutical compositions of the present invention include those that also contain one or more other active ingredients, in addition to a compound of the present invention.
The weight ratio of the compound of the compound of the present invention to the second active ingredient may be varied and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used. Thus, for example, when a compound of the present invention is combined with another agent, the weight ratio of the compound of the present invention to the other agent will generally range from about 1000:1 to about 1 :1000, such as about 200:1 to about 1 :200. Combinations of a compound of the present invention and other active ingredients will generally also be within the aforementioned range, but in each case, an effective dose of each active ingredient should be used. In such combinations the compound of the present invention and other active agents may be administered separately or in conjunction. In addition, the administration of one element may be prior to, concurrent to, or subsequent to the administration of other agent(s). The compounds of the present invention may be administered in conbination with other compounds which are known in the art to be useful for enhancing sleep quality and preventing and treating sleep disorders and sleep disturbances, including e.g., sedatives, hypnotics, anxiolytics, antipsychotics, antianxiety agents, antihistamines, benzodiazepines, barbiturates, cyclopyrrolones, GABA agonists, 5HT-2 antagonists including 5HT-2A antagonists and 5HT-2A/2C antagonists, histamine antagonists including histamine H3 antagonists, histamine H3 inverse agonists, imidazopyridines, minor tranquilizers, melatonin agonists and antagonists, melatonergic agents, other orexin antagonists, orexin agonists, prokineticin agonists and antagonists, pyrazolopyrimidines, T-type calcium channel antagonists, triazolopyridines, and the like, such as: adinazolam, allobarbital, alonimid, alprazolam, amitriptyline, amobarbital, amoxapine, armodafinil, APD-125, bentazepam, benzoctamine, brotizolam, bupropion, busprione, butabarbital, butalbital, capromorelin, capuride, carbocloral, chloral betaine, chloral hydrate, chlordiazepoxide, clomipramine, clonazepam, cloperidone, clorazepate, clorethate, clozapine, conazepam, cyprazepam, desipramine, dexclamol, diazepam, dichloralphenazone, divalproex, diphenhydramine, doxepin, EMD-281014, eplivanserin, estazolam, eszopiclone, ethchlorynol, etomidate, fenobam, flunitrazepam, flurazepam, fluvoxamine, fluoxetine, fosazepam, gaboxadol, glutethimide, halazepam, hydroxyzine, ibutamoren, imipramine, indiplon, lithium, lorazepam, lormetazepam, LY-156735, maprotiline, MDL-100907, mecloqualone, melatonin, mephobarbital, meprobamate, methaqualone, methyprylon, midaflur, midazolam, modafinil, nefazodone, NGD-2-73, nisobamate, nitrazepam, nortriptyline, ornortriptyline, oxazepam, paraldehyde, paroxetine, pentobarbital, perlapine, perphenazine, phenelzine, phenobarbital, prazepam, promethazine, propofol, protriptyline, quazepam, ramelteon, reclazepam, roletamide, secobarbital, sertraline, suproclone, TAK-375, temazepam, thioridazine, tiagabine, tracazolate, tranylcypromaine, trazodone, triazolam, trepipam, tricetamide, triclofos, trifluoperazine, trimetozine, trimipramine, uldazepam, venlafaxine, zaleplon, zolazepam, zopiclone, Zolpidem, and salts thereof, and combinations thereof, and the like, or the compound of the present invention may be administered in conjunction with the use of physical methods such as with light therapy or electrical stimulation.
In another embodiment, the subject compound may be employed in combination with other compounds which are known in the art, either administered separately or in the same pharmaceutical compositions, include, but are not limited to: insulin sensitizers including (i) PPARγ antagonists such as glitazones (e.g. ciglitazone; darglitazone; englitazone; isaglitazone (MCC-555); pioglitazone; rosiglitazone; troglitazone; tularik; BRL49653; CLX-0921; 5-BTZD), GW-0207, LG- 100641, and LY-300512, and the like); (iii) biguanides such as metformin and phenformin; (b) insulin or insulin mimetics, such as biota, LP-100, novarapid, insulin detemir, insulin lispro, insulin glargine, insulin zinc suspension (lente and ultralente); Lys-Pro insulin, GLP-I (73-7) (insulintropin); and GLP-I (7-36)-NH2); (c) sulfonylureas, such as acetohexamide; chlorpropamide; diabinese; glibenclamide; glipizide; glyburide; glimepiride; gliclazide; glipentide; gliquidone; glisolamide; tolazamide; and tolbutamide; (d) α-glucosidase inhibitors, such as acarbose, adiposine; camiglibose; emiglitate; miglitol; voglibose; pradimicin-Q; salbostatin; CKD-71 1 ; MDL-25,637; MDL-73,945; and MOR 14, and the like; (e) cholesterol lowering agents such as (i) HMG-CoA reductase inhibitors (atorvastatin, itavastatin, fluvastatin, lovastatin, pravastatin, rivastatin, rosuvastatin, simvastatin, and other statins), (ii) bile acid absorbers/sequestrants, such as cholestyramine, colestipol, dialkylaminoalkyl derivatives of a cross-linked dextran; Colestid®; LoCholest®, and the like, (ii) nicotinyl alcohol, nicotinic acid or a salt thereof, (iii) proliferator-activater receptor α agonists such as fenofibric acid derivatives (gemfibrozil, clofibrate, fenofibrate and benzafibrate), (iv) inhibitors of cholesterol absorption such as stanol esters, beta-sitosterol, sterol glycosides such as tiqueside; and azetidinones such as ezetimibe, and the like, and (acyl CoAxholesterol acyltransferase (ACAT)) inhibitors such as avasimibe, and melinamide, (v) anti-oxidants, such as probucol, (vi) vitamin E, and (vii) thyromimetics; (f) PP ARa agonists such as beclofibrate, benzafibrate, ciprofibrate, clofibrate, etofibrate, fenofibrate, and gemfibrozil; and other fibric acid derivatives, such as Atromid®, Lopid® and Tricor®, and the like, and PP ARa agonists as described in WO 97/36579; (g) PPARδ agonists, such as those disclosed in WO97/28149; (h) PPAR α/δ agonists, such as muraglitazar, and the compounds disclosed in US 6,414,002; (i) anti-obesity agents, such as (1) growth hormone secretagogues, growth hormone secretagogue receptor agonists/antagonists, such as NN703, hexarelin, MK-0677, SM-130686, CP-424,391, L-692,429, and L-163,255, and such as those disclosed in U.S. Patent Nos. 5,536,716, and 6,358,951, U.S. Patent Application Nos. 2002/049196 and 2002/022637, and PCT Application Nos. WO 01/56592 and WO 02/32888; (2) protein tyrosine phosphatase- IB (PTP-IB) inhibitors; (3) cannabinoid receptor ligands, such as cannabinoid CBi receptor antagonists or inverse agonists, such as rimonabant, taranabant, AMT-251, and SR- 14778 and SR 141716A (Sanofi Synthelabo), SLV-319 (Solvay), BAY 65-2520 (Bayer) and those disclosed in U.S. Patent Nos. 5,532,237, 4,973,587, 5,013,837, 5,081,122, 5,1 12,820, 5,292,736, 5,624,941, 6,028,084, PCT Application Nos. WO 96/33159, WO 98/33765, WO98/43636, WO98/43635, WO 01/09120, WO98/31227, WO98/41519, WO98/37061 , WO00/10967, WOOO/10968, WO97/29079, WO99/02499, WO 01/58869, WO 01/64632, WO 01/64633, WO 01/64634, W002/076949, WO 03/007887, WO 04/048317, and WO 05/000809; (4) anti-obesity serotonergic agents, such as fenfluramine, dexfenfluramine, phentermine, and sibutramine; (5) β3-adrenoreceptor agonists, such as AD9677/TAK677 (Dainippon/Takeda), CL-316,243, SB 418790, BRL-37344, L-796568, BMS- 196085, BRL- 35135A, CGP12177A, BTA-243, Trecadrine, Zeneca D71 14, SR 591 19A; (6) pancreatic lipase inhibitors, such as orlistat (Xenical®), Triton WR1339, RHC80267, lipstatin, tetrahydrolipstatin, teasaponin, diethylumbelliferyl phosphate, and those disclosed in PCT Application No. WO 01/77094; (7) neuropeptide Yl antagonists, such as BIBP3226, J-1 15814, BIBO 3304, LY- 357897, CP-671906, GI-264879A, and those disclosed in U.S. Patent No. 6,001,836, and PCT Patent Publication Nos. WO 96/14307, WO 01/23387, WO 99/51600, WO 01/85690, WO 01/85098, WO 01/85173, and WO 01/89528; (8) neuropeptide Y5 antagonists, such as GW- 569180A, GW-594884A, GW-587081X, GW-5481 18X, FR226928, FR 240662, FR252384, 1229U91 , GI-264879A, CGP71683A, LY-377897, PD-160170, SR-120562A, SR-120819A and JCF-104, and those disclosed in U.S. Patent Nos. 6,057,335; 6,043,246; 6,140,354; 6,166,038; 6,180,653; 6,191,160; 6,313,298; 6,335,345; 6,337,332; 6,326,375; 6,329,395; 6,340,683; 6,388,077; 6,462,053; 6,649,624; and 6,723,847, European Patent Nos. EP-01010691 , and EP- 01044970; and PCT International Patent Publication Nos. WO 97/19682, WO 97/20820, WO 97/20821, WO 97/20822, WO 97/20823, WO 98/24768; WO 98/25907; WO 98/25908; WO 98/27063, WO 98/47505; WO 98/40356; WO 99/15516; WO 99/27965; WO 00/64880, WO 00/68197, WO 00/69849, WO 01/09120, WO 01/14376; WO 01/85714, WO 01/85730, WO 01/07409, WO 01/02379, WO 01/02379, WO 01/23388, WO 01/23389, WO 01/44201, WO 01/62737, WO 01/62738, WO 01/09120, WO 02/22592, WO 0248152, and WO 02/49648; WO 02/094825; WO 03/014083; WO 03/10191; WO 03/092889; WO 04/002986; and WO 04/031175; (9) melanin-concentrating hormone (MCH) receptor antagonists, such as those disclosed in WO 01/21577 and WO 01/21 169; (10) melanin-concentrating hormone 1 receptor (MCHlR) antagonists, such as T-226296 (Takeda), and those disclosed in PCT Patent Application Nos. WO 01/82925, WO 01/87834, WO 02/051809, WO 02/06245, WO 02/076929, WO 02/076947, WO 02/04433, WO 02/51809, WO 02/083134, WO 02/094799, WO 03/004027; (11) melanin-concentrating hormone 2 receptor (MCH2R) agonist/antagonists; (12) orexin receptor antagonists, such as SB-334867-A, and those disclosed in patent publications herein; (13) serotonin reuptake inhibitors such as fluoxetine, paroxetine, and sertraline; (14) melanocortin agonists, such as Melanotan II; (15) Mc4r (melanocortin 4 receptor) agonists, such as CHIR86036 (Chiron), ME-10142, and ME-10145 (Melacure), CHIR86036 (Chiron); PT-141 , and PT-14 (Palatin); (16) 5HT-2 agonists; (17) 5HT2C (serotonin receptor 2C) agonists, such as BVT933, DPCA37215, WAY161503, R-1065, and those disclosed in U.S. Patent No. 3,914,250, and PCT Application Nos. WO 02/36596, WO 02/48124, WO 02/10169, WO 01/66548, WO 02/44152, WO 02/51844, WO 02/40456, and WO 02/40457; (18) galanin antagonists; (19) CCK agonists; (20) CCK-A (cholecystokinin-A) agonists, such as AR-R 15849, GI 181771, JMV- 180, A-71378, A-71623 and SR14613, and those discribed in U.S. Patent No. 5,739,106; (21) GLP-I agonists; (22) corticotropin-releasing hormone agonists; (23) histamine receptor-3 (H3) modulators; (24) histamine receptor-3 (H3) antagonists/inverse agonists, such as hioperamide, 3- (1H-imidazol-4-yl)propyl N-(4-pentenyl)carbamate, clobenpropit, iodophenpropit, imoproxifan, GT2394 (Gliatech), and O-[3-(1H-imidazol-4-yl)propanol]-carbamates; (25) β-hydroxy steroid dehydrogenase- 1 inhibitors (β-HSD-1); (26) PDE (phosphodiesterase) inhibitors, such as theophylline, pentoxifylline, zaprinast, sildenafil, amrinone, milrinone, cilostamide, rolipram, and cilomilast; (27) phosphodiesterase-3B (PDE3B) inhibitors; (28) NE (norepinephrine) transport inhibitors, such as GW 320659, despiramine, talsupram, and nomifensine; (29) ghrelin receptor antagonists, such as those disclosed in PCT Application Nos. WO 01/87335, and WO 02/08250; (30) leptin, including recombinant human leptin (PEG-OB, Hoffman La Roche) and recombinant methionyl human leptin (Amgen); (31) leptin derivatives; (32) BRS3 (bombesin receptor subtype 3) agonists. such as [D-Phe6,beta-Alal l,Phel 3,Nlel4]Bn(6-14) and [D-
Phe6,Phel3]Bn(6-13)propylamide, and those compounds disclosed in Pept. Sci. 2002 Aug; 8(8): 461-75); (33) CNTF (Ciliary neurotrophic factors), such as GI-181771 (Glaxo-SmithKline), SR 146131 (Sanofi Synthelabo), butabindide, PD 170,292, and PD 149164 (Pfizer); (34) CNTF derivatives, such as axokine (Regeneron); (35) monoamine reuptake inhibitors, such as sibutramine; (36) UCP-I (uncoupling protein-1), 2, or 3 activators, such as phytanic acid, 4-[(E)- 2-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-napthalenyl)-1-propenyl]benzoic acid (TTNPB), retinoic acid; (37) thyroid hormone β agonists, such as KB-261 1 (KaroBioBMS); (38) FAS (fatty acid synthase) inhibitors, such as Cerulenin and C75; (39) DGATl (diacylglycerol acyltransferase 1) inhibitors; (40) DGAT2 (diacylglycerol acyltransferase 2) inhibitors; (41) ACC2 (acetyl-CoA carboxylase-2) inhibitors; (42) glucocorticoid antagonists; (43) acyl- estrogens, such as oleoyl-estrone, disclosed in del Mar-Grasa, M. et al., Obesity Research, 9:202- 9 (2001); (44) dipeptidyl peptidase IV (DP-IV) inhibitors, such as isoleucine thiazolidide, valine pyrrolidide, NVP-DPP728, LAF237, P93/01, TSL 225, TMC-2A/2B/2C, FE 99901 1, P9310/K364, VIP 0177, SDZ 274-444, sitagliptin; and the compounds disclosed in US 6,699,871 , WO 03/004498; WO 03/004496; EP 1 258 476; WO 02/083128; WO 02/062764; WO 03/000250; WO 03/002530; WO 03/002531 ; WO 03/002553; WO 03/002593; WO 03/000180; and WO 03/000181; (46) dicarboxylate transporter inhibitors; (47) glucose transporter inhibitors; (48) phosphate transporter inhibitors; (49) Metformin (Glucophage®); (50) Topiramate (Topimax®); (50) peptide YY, PYY 3-36, peptide YY analogs, derivatives, and fragments such as BIM-43073D, BIM-43004C (Olitvak, D.A. et al., Dig. Dis. Sci. 44(3):643-48 (1999)); (51) Neuropeptide Y2 (NPY2) receptor agonists such NPY3-36, N acetyl [Leu(28,31)] NPY 24-36, TASP-V, and cyclo-(28/32)-Ac-[Lys28-Glu32]-(25-36)-pNPY; (52) Neuropeptide Y4 (NP Y4) agonists such as pancreatic peptide (PP), and other Y4 agonists such as 1229U91; (54) cyclooxygenase-2 inhibitors such as etoricoxib, celecoxib, valdecoxib, parecoxib, lumiracoxib, BMS347070, tiracoxib or JTE522, ABT963, CS502 and GW406381 ; (55)
Neuropeptide Yl (NPYl) antagonists such as BIBP3226, J-115814, BIBO 3304, LY-357897, CP-671906, GI-264879A; (56) Opioid antagonists such as nalmefene (Revex ®), 3- methoxynaltrexone, naloxone, naltrexone; (57) 1 lβ HSD-I (11 -beta hydroxy steroid dehydrogenase type 1) inhibitors such as BVT 3498, BVT 2733, and those disclosed in WO 01/90091, WO 01/90090, WO 01/90092, US 6,730,690 and US 2004-013301 1; (58) aminorex; (59) amphechloral; (60) amphetamine; (61) benzphetamine; (62) chlorphentermine; (63) clobenzorex; (64) cloforex; (65) clominorex; (66) clortermine; (67) cyclexedrine; (68) dextroamphetamine; (69) diphemethoxidine, (70) N-ethylamphetamine; (71) fenbutrazate; (72) fenisorex; (73) fenproporex; (74) fludorex; (75) fluminorex; (76) furfurylmethylamphetamine; (77) levamfetamine; (78) levophacetoperane; (79) mefenorex; (80) metamfepramone; (81) methamphetamine; (82) norpseudoephedrine; (83) pentorex; (84) phendimetrazine; (85) phenmetrazine; (86) picilorex; (87) phytopharm 57; and (88) zonisamide., (89) neuromedin U and analogs or derivatives thereof, (90) oxyntomodulin and analogs or derivatives thereof, and (91) Neurokinin- 1 receptor antagonists (NK-I antagonists) such as the compounds disclosed in: U.S. Patent Nos. 5,162,339, 5,232,929, 5,242,930, 5,373,003, 5,387,595, 5,459,270, 5,494,926, 5,496,833, and 5,637,699. In another embodiment, the subject compound may be employed in combination with an anti-depressant or anti-anxiety agent, including norepinephrine reuptake inhibitors (including tertiary amine tricyclics and secondary amine tricyclics), selective serotonin reuptake inhibitors (SSRIs), monoamine oxidase inhibitors (MAOIs), reversible inhibitors of monoamine oxidase (RIMAs), serotonin and noradrenaline reuptake inhibitors (SNRIs), corticotropin releasing factor (CRF) antagonists, α-adrenoreceptor antagonists, neurokinin- 1 receptor antagonists, atypical anti-depressants, benzodiazepines, 5-HTI A agonists or antagonists, especially 5-HTI A partial agonists, and corticotropin releasing factor (CRF) antagonists. Specific agents include: amitriptyline, clomipramine, doxepin, imipramine and trimipramine; amoxapine, desipramine, maprotiline, nortriptyline and protriptyline; citalopram, duloxetine, fluoxetine, fluvoxamine, paroxetine and sertraline; isocarboxazid, phenelzine, tranylcypromine and selegiline; moclobemide: venlafaxine; aprepitant; bupropion, lithium, nefazodone, trazodone and viloxazine; alprazolam, chlordiazepoxide, clonazepam, chlorazepate, diazepam, halazepam, lorazepam, oxazepam and prazepam; buspirone, flesinoxan, gepirone and ipsapirone, and pharmaceutically acceptable salts thereof. In another embodiment, the subject compound may be employed in combination with anti- Alzheimer's agents; beta-secretase inhibitors; gamma-secretase inhibitors; growth hormone secretagogues; recombinant growth hormone; HMG-CoA reductase inhibitors; NS AID's including ibuprofen; vitamin E; anti-amyloid antibodies; CB-I receptor antagonists or CB-I receptor inverse agonists; antibiotics such as doxycycline and rifampin; N-methyl-D- aspartate (NMDA) receptor antagonists, such as memantine; cholinesterase inhibitors such as galantamine, rivastigmine, donepezil, and tacrine; growth hormone secretagogues such as ibutamoren, ibutamoren mesylate, and capromorelin; histamine H3 antagonists; AMPA agonists; PDE IV inhibitors; GABAA inverse agonists; or neuronal nicotinic agonists.
In another embodiment, the subject compound may be employed in combination with sedatives, hypnotics, anxiolytics, antipsychotics, antianxiety agents, cyclopyrrolones, imidazopyridines, pyrazolopyrimidines, minor tranquilizers, melatonin agonists and antagonists, melatonergic agents, benzodiazepines, barbiturates, 5HT-2 antagonists, and the like, such as: adinazolam, allobarbital, alonimid, alprazolam, amitriptyline, amobarbital, amoxapine, bentazepam, benzoctamine, brotizolam, bupropion, busprione, butabarbital, butalbital, capuride, carbocloral, chloral betaine, chloral hydrate, chlordiazepoxide, clomipramine, clonazepam, cloperidone, clorazepate, clorethate, clozapine, cyprazepam, desipramine, dexclamol, diazepam, dichloralphenazone, divalproex, diphenhydramine, doxepin, estazolam, ethchlorvynol, etomidate, fenobam, flunitrazepam, flurazepam, fluvoxamine, fluoxetine, fosazepam, glutethimide, halazepam, hydroxyzine, imipramine, lithium, lorazepam, lormetazepam, maprotiline, mecloqualone, melatonin, mephobarbital, meprobamate, methaqualone, midaflur, midazolam, nefazodone, nisobamate, nitrazepam, nortriptyline, oxazepam, paraldehyde, paroxetine, pentobarbital, perlapine, perphenazine, phenelzine, phenobarbital, prazepam, promethazine, propofol, protriptyline, quazepam, reclazepam, roletamide, secobarbital, sertraline, suproclone, temazepam, thioridazine, tracazolate, tranylcypromaine, trazodone, triazolam, trepipam, tricetamide, triclofos, trifluoperazine, trimetozine, trimipramine, uldazepam, venlafaxine, zaleplon, zolazepam, Zolpidem, and salts thereof, and combinations thereof, and the like, or the subject compound may be administered in conjunction with the use of physical methods such as with light therapy or electrical stimulation.
In another embodiment, the subject compound may be employed in combination with levodopa (with or without a selective extracerebral decarboxylase inhibitor such as carbidopa or benserazide), anticholinergics such as biperiden (optionally as its hydrochloride or lactate salt) and trihexyphenidyl (benzhexol) hydrochloride, COMT inhibitors such as entacapone, MOA-B inhibitors, antioxidants, A2a adenosine receptor antagonists, cholinergic agonists, NMDA receptor antagonists, serotonin receptor antagonists and dopamine receptor agonists such as alentemol, bromocriptine, fenoldopam, lisuride, naxagolide, pergolide and pramipexole.
In another embodiment, the subject compound may be employed in combination with acetophenazine, alentemol, benzhexol, bromocriptine, biperiden, chlorpromazine, chlorprothixene, clozapine, diazepam, fenoldopam, fluphenazine, haloperidol, levodopa, levodopa with benserazide, levodopa with carbidopa, lisuride, loxapine, mesoridazine, molindolone, naxagolide, olanzapine, pergolide, perphenazine, pimozide, pramipexole, risperidone, sulpiride, tetrabenazine, trihexyphenidyl, thioridazine, thiothixene or trifluoperazine. In another embodiment, the subject compound may be employed in combination with a compound from the phenothiazine, thioxanthene, heterocyclic dibenzazepine, butyrophenone, diphenylbutylpiperidine and indolone classes of neuroleptic agent. Suitable examples of phenothiazines include chlorpromazine, mesoridazine, thioridazine, acetophenazine, fluphenazine, perphenazine and trifluoperazine. Suitable examples of thioxanthenes include chlorprothixene and thiothixene. An example of a dibenzazepine is clozapine. An example of a butyrophenone is haloperidol. An example of a diphenylbutylpiperidine is pimozide. An example of an indolone is molindolone. Other neuroleptic agents include loxapine, sulpiride and risperidone. In another embodiment, the subject compound may be employed in combination with a nicotine agonist or a nicotine receptor partial agonist such as varenicline, opioid antagonists (e.g., naltrexone (including naltrexone depot), antabuse, and nalmefene), dopaminergic agents (e.g., apomorphine), ADD/ ADHD agents (e.g., methylphenidate hydrochloride (e.g., R1talin® and Concerta®), atomoxetine (e.g., Strattera®), a monoamine oxidase inhibitor (MAOI), amphetamines (e.g., Adderall®)) and anti-obesity agents, such as apo- B/MTP inhibitors, 1 1 Beta-hydroxy steroid dehydrogenase- 1 (1 IBeta-HSD type 1) inhibitors, peptide YY3-36 or analogs thereof, MCR-4 agonists, CCK-A agonists, monoamine reuptake inhibitors, sympathomimetic agents, β3 adrenergic receptor agonists, dopamine receptor agonists, melanocyte-stimulating hormone receptor analogs, 5-HT2c receptor agonists, melanin concentrating hormone receptor antagonists, leptin, leptin analogs, leptin receptor agonists, galanin receptor antagonists, lipase inhibitors, bombesin receptor agonists, neuropeptide-Y receptor antagonists (e.g., NPY Y5 receptor antagonists), thyromimetic agents, dehydroepiandrosterone or analogs thereof, glucocorticoid receptor antagonists, other orexin receptor antagonists, glucagon-like peptide- 1 receptor agonists, ciliary neurotrophic factors, human agouti-related protein antagonists, ghrelin receptor antagonists, histamine 3 receptor antagonists or inverse agonists, and neuromedin U receptor agonists, and pharmaceutically acceptble salts thereof.
In another embodiment, the subject compound may be employed in combination with an anoretic agent such as aminorex, amphechloral, amphetamine, benzphetamine, chlorphentermine, clobenzorex, cloforex, clominorex, clortermine, cyclexedrine, dexfenfluramine, dextroamphetamine, diethylpropion, diphemethoxidine, N-ethylamphetamine, fenbutrazate, fenfluramine, fenisorex, fenproporex, fludorex, fluminorex, furfurylmethylamphetamine, levamfetamine, levophacetoperane, mazindol, mefenorex, metamfepramone, methamphetamine, norpseudoephedrine, pentorex, phendimetrazine, phenmetrazine, phentermine, phenylpropanolamine, picilorex and sibutramine; selective serotonin reuptake inhibitor (SSRI); halogenated amphetamine derivatives, including chlorphentermine, cloforex, clortermine, dexfenfluramine, fenfluramine, picilorex and sibutramine; and pharmaceutically acceptble salts thereof.
In another embodiment, the subject compound may be employed in combination with an opiate agonist, a lipoxygenase inhibitor, such as an inhibitor of 5-lipoxygenase, a cyclooxygenase inhibitor, such as a cyclooxygenase-2 inhibitor, an interleukin inhibitor, such as an interleukin- 1 inhibitor, an NMDA antagonist, an inhibitor of nitric oxide or an inhibitor of the synthesis of nitric oxide, a non-steroidal antiinflammatory agent, or a cytokine-suppressing antiinflammatory agent, for example with a compound such as acetaminophen, asprin, codiene, fentanyl, ibuprofen, indomethacin, ketorolac, morphine, naproxen, phenacetin, piroxicam, a steroidal analgesic, sufentanyl, sunlindac, tenidap, and the like. Similarly, the subject compound may be administered with a pain reliever; a potentiator such as caffeine, an H2-antagonist, simethicone, aluminum or magnesium hydroxide; a decongestant such as phenylephrine, phenylpropanolamine, pseudophedrine, oxymetazoline, epinephrine, naphazoline, xylometazoline, propylhexedrine, or levo-desoxy-ephedrine; an antiitussive such as codeine, hydrocodone, caramiphen, carbetapentane, or dextramethorphan; a diuretic; and a sedating or non-sedating antihistamine.
The compounds of the present invention may be administered by oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, ICV, intracisternal injection or infusion, subcutaneous injection, or implant), by inhalation spray, nasal, vaginal, rectal, sublingual, or topical routes of administration and may be formulated, alone or together, in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles appropriate for each route of administration. In addition to the treatment of warmblooded animals such as mice, rats, horses, cattle, sheep, dogs, cats, monkeys, etc., the compounds of the invention are effective for use in humans. The pharmaceutical compositions for the administration of the compounds of this invention may conveniently be presented in dosage unit form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the active ingredient into association with the carrier which constitutes one or more accessory ingredients. In general, the pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation. In the pharmaceutical composition the active object compound is included in an amount sufficient to produce the desired effect upon the process or condition of diseases. As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. Pharmaceutical compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. Compositions for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil. Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Oily suspensions may be formulated by suspending the active ingredient in a suitable oil. Oil-in-water emulsions may also be employed. Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Pharmaceutical compositions of the present compounds may be in the form of a sterile injectable aqueous or oleagenous suspension. The compounds of the present invention may also be administered in the form of suppositories for rectal administration. For topical use, creams, ointments, jellies, solutions or suspensions, etc., containing the compounds of the present invention may be employed. The compounds of the present invention may also be formulated for administered by inhalation. The compounds of the present invention may also be administered by a transdermal patch by methods known in the art.
Several methods for preparing the compounds of this invention are illustrated in the following Schemes and Examples. Starting materials are made according to procedures known in the art or as illustrated herein. The following abbreviations are used herein: Me: methyl; Et: ethyl; t-Bu: tert-butyl; Ar: aryl; Ph: phenyl; Bn: benzyl; Ac: acetyl; ACN: acetonitrile; THF: tetrahydrofuran; DEAD: diethylazodicarboxylate; DIPEA: N,N- diisopropylethylamine; NMM: N-methylmorpholine; DMSO: dimethylsulfoxide; EDC: N-(3- Dimethylaminopropyl)-N'-ethylcarbodiimide; HOBT: hydroxybenzotriazole hydrate; Boc: tert- butyloxy carbonyl; Et3N: triethylamine; DCM: dichloromethane; DCE: dichloroethane; BSA: bovine serum albumin; TFA: trifluoracetic acid; DMF: N,N-dimethylformamide; MTBE: methyl tert-butyl ether; SOCl2: thionyl chloride; CDI: carbonyl diimidazole; PyCIu: l-(chloro-1- pyrrolidinylmethylene)-pyrrolidinium hexafluorophosphate; T3P: 1-propylphosphonic acid cyclic anhydride; rt: room temperature; HPLC: high performance liquid chromatography. The compounds of the present invention can be prepared in a variety of fashions.
In some cases the final product may be further modified, for example, by manipulation of substituents. These manipulations may include, but are not limited to, reduction, oxidation, alkylation, acylation, and hydrolysis reactions which are commonly known to those skilled in the art. In some cases the order of carrying out the foregoing reaction schemes may be varied to facilitate the reaction or to avoid unwanted reaction products. The following examples are provided so that the invention might be more fully understood. These examples are illustrative only and should not be construed as limiting the invention in any way.
SCHEME A
Figure imgf000039_0001
5-Aryl-2-chloronicotinic acids (A-2) are synthesized via Suzuki coupling reactions on A-I with aryl boronic under standard conditions followed by hydrolysis. A-2 are coupled to amines using PyCIu to give A-3. Coupling of A-3 with boronic acids using standard Suzuki coupling conditions under microwave heating affords A-4.
SCHEME B
Figure imgf000040_0001
2,5-Disubstituted nicotinic acids (B-2) can be synthesized via B-I by selective Suzuki coupling on the 5-position followed by another Suzuki coupling on the 2-position and in situ hydrolysis. Compounds such as B^2 can be advanced to final compounds through amide coupling to afford B-3.
SCHEME C
Figure imgf000040_0002
2,5-Disubstituted nicotinic acids (C-2) can be synthesized via C-I by selective Suzuki coupling on the 5-position followed by another Suzuki coupling on the 2-position followed by basic hydrolysis. Compounds such as C-2 can be advanced to final compounds through amide coupling to afford C-3.
SCHEME D
Figure imgf000041_0001
2,5-Disubstituted nicotinic acids (D-2) can be synthesized via D-I by selective Suzuki coupling on the 5-position followed by nucleophilic substitution on the 2-position and hydrolysis of the ester. Compounds such as D-2 can be advanced to final compounds through amide coupling to afford D-3.
SCHEME E
Figure imgf000041_0002
2,5-Disubstituted nicotinic acids ( E-2) can be synthesized via E-1 by selective Suzuki coupling on the 5-position followed by nucleophilic substitution with N-H containing heterocycles on the 2-position. Hydrolysis of the ester affords E-2 Compounds such as E-2 can be advanced to final compounds through amide coupling to afford E-3.
SCHEME F
Figure imgf000042_0001
2,5-Disubstituted nicotinic acids ( F-2) can be synthesized via IM. by selective Suzuki coupling on the 5-position followed by Negishi coupling on the 2-position. Hydrolysis of the ester affords F-2. Compounds such as F-2 can be advanced to final compounds through amide coupling to afford F-3.
SCHEME G
Figure imgf000042_0002
2,5-Disubstituted nicotinic esters (G-2) can be synthesized via G-I by selective Stille coupling on the 5-position followed by another Stille coupling on the 2-position. Compounds such as G-2 can be advanced to final compounds through transamination mediated by trimethylaluminum to afford G-3.
SCHEME H
Figure imgf000043_0001
2,5-Disubstituted nicotinic acids (H-2) can be synthesized via F-I by selective Suzuki coupling on the 5-position followed by Stille coupling on the 2-position. Hydrolysis of the ester affords H-2. Compounds such as H-2 can be advanced to final compounds through amide coupling to afford H-3.
EXAMPLE 1
Figure imgf000043_0002
2-ChIQrQ-5-(3, 5-dimethylphenyl)nicotinic acid (1-2)
To a deoxygenated solution of methyl 2-chloro-5-iodonicotinate (IdL 2.01 g, 6.72 mmol, 1 equiv) and 3,5-dimethylphenylboronic acid (1.01 g, 6.72 mmol. 1.00 equiv) in dioxane (250 mL) was added tetrakis(triphenylphosphine)palladium(0) (155 mg, 0.134 mmol) and 2 N aqueous sodium bicarbonate solution (16.8 mL, 33.6 mmol, 5.00 equiv) and the reaction mixture was heated at 90 °C for 48 hours. The reaction mixture was allowed to cool to 23 ° C, then partitioned between ethyl acetate (500 mL) and saturated aqueous sodium bicarbonate solution (500 mL). The aqueous layer was filtered and acidified to pH 2 with concentrated hydrochloric acid. The resulting precipitate was collected by filtration washed with water (2 x 100 mL) and air dried to afford 2-chloro-5-(3,5-dimethylphenyl)nicotinic acid (1-2) as an off white solid. LRMS m/z (M+H) 262.3 found, 262.1 required.
2-Chloro-N-(3,4-dimethoxybenzyl)-5-(3.5-dimethylphenyl)nicotinamide (1-3)
To a solution of 2-chloro-5-(3,5-dimethylphenyl)nicotinic acid (1-2, 0.200 g, 0.764 mmol, 1 equiv) and 3,4-dimethoxybenzylamine (0.319g, 1.91 mmol, 2.50 equiv) in DMF (5 mL) was added l-(chloro-1-pyrrolidinylmethylene)-pyrrolidinium hexafluorophosphate (PyCIu, 0.383 mg, 1.15 mmol, 1.51 equiv), and diisopropylethylamine (0.667 mL, 3.82 mmol, 5.00 equiv). The resulting mixture was stirred at 23 ° C for 20 hours. The reaction mixture was filtered and purified via reverse phase liquid chromatography (H2O/CH3CN gradient w/ 0.1 % TFA present). The desired fractions were partitioned between saturated aqueous sodium bicarbonate solution and ethyl acetate (2 x 50 mL). The combined organic layers were washed with brine then dried over sodium sulfate and concentrated to give 2-chloro-jV-(3,4- dimethoxybenzyl)-5-(3,5-dimethylphenyl)nicotinamide (1-3) as a white powder. LRMS m/z (M+H) 262.3 found, 262.1 required.
N-(3,4-Dimethoxybenzyl)-5-(3,5-dimethylphenyl)-2-(l-methyl- 1H-pyrazol-4-yl)nicotinamide (1-
4)
To a solution of 2-chloro-N-(3,4-dimethoxybenzyl)-5-(3,5- dimethylphenyl)nicotinamide (1-3, 50.0 mg, 0.122 mmol)) and l-methyl-4-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)-1H-pyrazole (25.3 mg, 0.122 mmol) in dioxanes (3 mL) was added tetrakis(triphenylphosphine)palladium(0) (7.03 mg, 6.08 μmol) and 2 N aqueous sodium bicarbonate solution (0.304 mL, 0.608 mmol). Under microwave heating the deoxygenated mixture was held at 160 °C for 1 h. The reaction mixture was filtered and concentrated to dryness. A solid residue was triturated with ethyl acetate (4 mL), collected by filtration, washed with water (10 mL) and air dried to afford N-(3,4-dimethoxybenzyl)-5-(3,5-dimethylphenyl)-2- (1 -methyl- 1H-pyrazol-4-yl)nicotinamide (1-4). 1H NMR (300 MHz, DMSO) δ 9.09 (t, 1H, J = 5.8 Hz), 8.87 (d, 1H, J= 1.8 Hz ), 7.92 (d, 1H, J= 1.8 Hz), 7.82 (d, 1H, J= 9.8 Hz), 7.39 (s, 2H), 7.06 (s, 1H), 6.92 (m, 3H), 4.29 (d, 2H, J= 5.3 Hz), 3.74 (m, 6H), 3.32 (s, 3H), 2.35 (s, 3H). HRMS m/z (M+H) 457.2242 found, 457.2234 required.
TABLE 1
The following compounds were prepared using the foregoing methodology, but substituting the appropriately substituted reagent, as described in the foregoing Reaction
Schemes and Examples. The requisite starting materials were commercially available, described in the literature or readily synthesized by one skilled in the art of organic synthesis without undue experimentation.
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
EXAMPLE 2
Figure imgf000049_0001
5-(3-Chloro-5-methvIphenyl)-2-(l -methyl- 1H-pyrazol-4-vQ nicotinic acid (2-2)
To a solution of methyl 2-chloro-5-iodonicotonate {2Λ_, 1.00 g, 3.36 mmol) in dimethylformamide (15 mL) at 25°C was added 3-chloro-5-methylboronic acid (0.573 g, 3.36 mmol; synthesized via procedures found in Org. Lett. 2007, P, 757-760), PdCl2dppf (0.246 g, 0.336 mmol) followed by IM aqueous cesium carbonate (13.5 mL, 13.5 mmol) and the system was stirred for 4h at 25°C. The system was partitioned between water and EtOAc, and dried over magnesium sulfate. Filtration and concentration yielded a brown solid which upon tritiration with ether afforded a tan solid. To this tan solid (0.05 g, 0.169 mmol) in dimethylformamide (0.8 mL) at 25°C was added l-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H pyrazole (0.038 g, 0.186 mmol), PdCl2dppf (0.012 g, 0.017 mmol) followed by 4M aqueous cesium carbonate (0.67 mL, 0.675 mmol) and the system was stirred for 15 minutes at 135°C in the microwave. The system was partitioned between water and EtOAc. The aqueous layer was then acidified using 25% citric acid to a pH of 3, extracted with EtOAc and the organic layer was dried over magnesium sulfate. Filtration and concentration afforded the title compound (2-2) as a cream solid. ESI+ MS [M+H]+ C17H14ClN3O2 = 328.0.
5-(3-Chloro-5-methylphenvn-N-(3.4-dimethoxybenzyl)-2-(l-methyl-1H-Dyrazol-4-vn nicotinamide (2-3) To a solution of 5-(3-chloro-5-methylphenyl)-2-(l -methyl- 1 H-pyrazol-4-yl) nicotinic acid (2-2, 0.050 g, 0.153 mmol) and 3,4-dimethoxybenzylamine (0.099g, 0.595 mmol) in dimethylformamide (1 mL) was added EDC (0.132 g, 0.686 mmol), HOBt (0.105 g, 0.686 mmol) followed by N-methylmorpholine (0.250 mL, 2.28 mmol) and the system was stirred at 25°C overnight. The reaction mixture was partitioned between ethyl acetate and water, washed with saturated sodium carbonate and dried over magnesium sulfate. The reaction mixture was filtered and purified via normal phase chromatography (0→5% MeOH in DCM) to afford the title compound (2=3) as a bone-colored powder. 1H NMR (500 MHz, CDCl3) δ 8.81 (d, J= 2.5Hz, 1H), 7.99 (s, 1H), 7.92 (d, J= 2.5Hz, 1H), 7.78 (s, 1H), 7.38 (s, 1H), 7.28 (m, 2H), 7.23 (s, 1H), 6.84 (s, 2H), 6.06 (m, 1H), 4.55 (d, J= 6Hz, 2H), 3.88(s, 3H), 3.87(s, 3H), 3.86(s, 3H), 2.42 (s, 3H). HRMS [M+H] C26H25ClN4O3 calc'd 477.1688, found 477.1682.
TABLE 2
The following compounds were prepared using the foregoing methodology, but substituting the appropriately substituted reagent, as described in the foregoing Reaction
Schemes and Examples. The requisite starting materials were commercially available, described in the literature or readily synthesized by one skilled in the art of organic synthesis without undue experimentation.
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0002
EXAMPLE 3
Figure imgf000053_0001
Methyl 6-chloro-5'-methyl-3,3'-bipyridine-5-carboxylate (3-2)
To a solution of methyl 2-chloro-5-iodonicotonate (3-1, 22g, 74.0 mmol) in dimethylformamide (296 mL) at 0°C (ice bath) was added 3-methyl-5-pyridylboronic acid (10.13 g, 74.0 mmol), PdChdppf (5.41 g, 7.40 mmol) followed by cesium carbonate (84 g, 259 mmol) and water (13.32 mL, 740 mmol) and the system was stirred overnight as the ice bath warmed to room temperature. The system was partitioned between water and EtOAc, extracted 3x with EtOAc, combined organics were washed with brine and dried over magnesium sulfate. Filtration and concentration yielded a brown oil which upon purification via normal phase chromatography (0→100% EtOAc in Hx) afforded the title compound (3-2) as an off-white solid. ESI+ MS [M+H]+ C13H11ClN2O2 = 262.9.
Methyl 6-(2-fluorophenyl)-5'-methyl-3,3'-bipyridine-5-carboxylate (3-3)
To a solution of methyl 6-chloro-5'-methyl-3,3'-bipyridine-5-carboxylate (3-2, 4.25 g, 16.16 mmol) in degassed dimethylformamide (81 mL) at 25°C was added 2- fluorophenylboronic acid (2.94 g, 21.01 mmol), PdCl2dppf (1.18 g, 1.62 mmol) and 4M aqueous cesium carbonate (10.1 mL, 40.4 mmol). The reaction flask was purged with nitrogen, sealed and stirred for 4 hours at 40°C. The crude reaction mixture was partitioned between water and EtOAc, separating layers and washing the organic layer with saturated sodium bicarbonate solution and brine. The solvent was removed by rotary evaporation and the crude product was taken up in DCM (40 mL). Quadrapure TU (6 g) was added and the mixture was stirred overnight at room temperature. Filtration and concentration afforded the title compound (3-3) as a solid. ESI+ MS [M+H]+ Ci9H15FN2O2 = 323.1.
6-(2-fluorophenyl)-5'-methyl-3,3'-bipyridine-5-carboxylic acid hydrochloride (3-4)
To a solution of methyl 6-(2-fluorophenyl)-5'-methyl-3,3'-bipyridine-5- carboxylate (3-3, 5.57 g, 17.28 mmol) in THF/water (84 mL; 5:1) was added 1OM sodium hydroxide solution (3.46 mL, 34.6 mmol) and the reaction was stirred at 60°C overnight. The reaction mixture was diluted with water, filtered and concentrated. The crude product was taken up in DMF/DMSO/CH3CN/water (72 mL;2:2: 1 :0.5), filtered and purified by reverse phase preparative chromatography (5-40% acetonitrile in water with 0.1% TFA buffer). The clean fractions were pooled and evaporated to give the TFA salt as a white solid. This material was taken up in a minimal amount of THF/acetonitrile and ethyl ether saturated with HCl was added, forming a white precipitate. The solvent was decanted away after centrifugation of the suspension. This was repeated 2 more times, once with HCl/ether and then with diethyl ether.
After the final decant, the solids were dried under high vacuum to afford the title compound (3-4) as the HCl salt. ESI+ MS [M+H]+ C18H13FN2O2 HCl = 309.1. N-[(5.6-dimethoxypyridin-2-yl)methyl]-6-(2-fluorophenyl)-5'-methyl-3,3'-bipyridine-5- carboxamide (3-5)
To a solution of 6-(2-fluorophenyl)-5'-methyl-3,3'-bipyridine-5-carboxylic acid hydrochloride (34, 3.0 g, 8.70 mmol) in dimethylformamide (43 mL) cooled to O °C , was added EDC (2.67 g, 13.92 mmol) and HOBt (2.13 g, 13.92 mmol) followed by l-(5,6- dimethoxypyridin-2-yl)methanamine (11-6, 1.9Og, 11.31 mmol) and N-methylmorpholine (5.74 mL, 52.2 mmol). The reaction was allowed to warm to room temperature and stir overnight. The reaction mixture was partitioned between ethyl acetate and saturated sodium bicarbonate. The organics were washed with water and brine, then stripped to dryness. The crude product was taken up in DMF/DMSO (30 mL, 1 :1), filtered and purified via reverse phase chromatography (5→65% acetonitrile in water with 0.1% TFA buffer). The clean fractions were pooled, partitioned between ethyl acetate and 2M sodium carbonate. The organic layer was washed with water and brine, dried over anhydrous sodium sulfate, filtered and concentrated to afford the title compound (3-5) as a white solid. 1H NMR (500 MHz, CDCl3) δ 8.99 (d, J= 2.5Hz, 1H), 8.74 (d, J= 2Hz, 1H) 8.53 (s, 1H), 8.34 (d, J= 2.5Hz, 1H), 7.77 (s, 1H), 7.66 (m, 1H), 7.31 (m, 1H), 7.21 (t, J=14.6 Hz, 1H), 6.98 (d, J=7.8 Hz, 1H), 6.92 (t, J=I 8.3 Hz, 1H), 6.73 (d, J=7.8 Hz, 1H), 6.46 (m, 1H), 4.44 (d, J= 5Hz, 2H), 3.87 (s, 3H), 3.81(s, 3H), 2.45 (s, 3H). HRMS [M+H] C26H23FN4O3 calc'd 459.1827, found 459.1830.
EXAMPLE 3A
Figure imgf000055_0001
N-[(5,6-dimethoxypyridin-2-yl)methyll-6-(3-fluorophenyl)-5'-methyl-3,3'-bipyridine-5- carboxamide (3-7)
To a solution of 6-(3-fluorophenyl)-5'-methyl-3,3'-bipyridine-5-carboxylic acid hydrochloride (3-6, prepared by the method described above for the preparation of 3-4, 2.5 g, 7.25 mmol) in dimethylformamide (36 mL) cooled to O °C , was added EDC (1.95 g, 10.2 mmol) and HOBt (1.67 g, 10.9 mmol) followed by l-(5,6-dimethoxypyridin-2-yl)methanamine (11-6, 1.46g, 8.7 mmol) and N-methylmorpholine (4.78 mL, 43.5 mmol). The reaction stirred for 15 minutes and then was allowed to warm to room temperature. After 3.5 hours the reaction mixture was partitioned between ethyl acetate and saturated sodium bicarbonate. The organics were washed with water and brine, dried over Na2SO4ZMgSO4, filtered and stripped to dryness. The crude product was recrystallized from chloroform/ ethyl ether to afford the title compound (3-7) as a white solid. 1H NMR (500 MHz, CDCl3) δ 8.96 (d, J= 2.2 Hz, 1H), 8.72 (d, J= 1.7 Hz, 1H), 8.56 (s, 1H), 8.23 (d, J= 2.2 Hz, 1H), 7.76 (s, 1H), 7.44 (d, J= 7.8 Hz, 1H), 7.40 (d, J= 9.5 Hz, 1H), 7.24 (m, 1H), 6.99 (m, 2H), 6.74 (d, J=7.8 Hz, 1H), 6.33 (m, 1H), 4.44 (d, J= 5.4 Hz, 2H), 3.88 (s, 3H), 3.76(s, 3H), 2.45 (s, 3H). HRMS [M+H] C26H23FN4O3 calc'd 459.1827, found 459.1832.
TABLE 3
The following compounds were prepared using the foregoing methodology, but substituting the appropriately substituted reagent, as described in the foregoing Reaction Schemes and Examples. The requisite starting materials were commercially available, described in the literature or readily synthesized by one skilled in the art of organic synthesis without undue experimentation.
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0002
Figure imgf000058_0001
5- -chloro-5-methylphenyl)-2-morpholin-4-ylnicotinic acid (4-2)
To a solution of methyl 2-chloro-5-iodonicotonate (4-1, 9.34 g, 31.5 mmol) in dimethylformamide (157 mL) at 25°C was added 3-chloro-5-methylboronic acid (5.35 g, 31.4 mmol), PdCl2dppf (2.3 g, 3.14 mmol) followed by cesium carbonate (41.0 g, 126 mmol) and water (10.0 mL, 565 mmol) and the system was stirred for 4 h at 25°C. The system was partitioned between water and EtOAc, and dried over magnesium sulfate. Filtration and concentration yielded a brown solid which upon purification via normal phase chromatography (0-→15% EtOAc in Hx) afforded a white solid. To this white solid (4.0 g, 13.5 mmol) in dioxane (67.5 mL) at 25°C was added morpholine (5.88 g, 67.5 mmol) and the system was heated to 90°C for 3h. The solvent was removed in vacuo and the resulting residue was partitioned between water and EtOAc followed by purification via normal phase chromatography (5— ->15% EtOAc in Hx) to yield a white foam. To this white foam (4.2 g, 12.11 mmol) in THF (30 mL) and MeOH (30 mL) was added KOH (5.19 mL, 36.3 mmol) and stirred at ambient temperature overnight. The system was then acidified using 6 N HCl to a pH of 2.0 and the solvents were azeotroped off with toluene to afford the title compound (4^) as a yellow powder. HRMS [M+H] Ci7Hi2ClN2O3 calc'd 333.1002, found 333.1000.
5-(3-chloro-5-methylphenyl)-N-(3,4-dimethoxybenzvD-2-(morpholin-4-yl) nicotinamide (4-3)
To a solution of 5-(3-chloro-5-methylphenyl)-2-rnorpholin-4-ylnicotinic acid (4-2, 2.8 g, 8.41 mmol) and 3,4-dimethoxybenzylamine (6.30 mL, 42.1 mmol) in DCM (42 mL) was added EDC (4.84 g, 25.2 mmol), HOBt (3.87 g, 25.2 mmol) followed by N-methylmorpholine (9.25 mL, 84 mmol) and the system was stirred at 25°C overnight. The reaction mixture was partitioned between ethyl acetate and water, washed with saturated sodium bicarbonate and dried over magnesium sulfate. The reaction mixture was filtered, concentrated and triturated with diethylether to afford the title compound (4-3) as a bone powder. 1H NMR (400 MHz, CDCl3) δ 8.82 (m, 1H), 8.53 (d, J= 2.4Hz, 1H), 8.49 (d, J= 2.4Hz, 1H), 7.35 (s, 1H), 7.16 (s, 1H), 6.90 (m, 2H), 6.84 (m, 1H), 4.56 (d, J= 5.6Hz, 2H), 3.86 (s, 3H), 3.85 (s, 3H), 3.55 (m, 4H), 3.12 (m, 4H), 2.37(s, 3H). HRMS [M+H] C26H28ClN3O4 calc'd 482.1841, found 482.1847.
TABLE 4
The following compounds were prepared using the foregoing methodology, but substituting the appropriately substituted reagent, as described in the foregoing Reaction
Schemes and Examples. The requisite starting materials were commercially available, described in the literature or readily synthesized by one skilled in the art of organic synthesis without undue experimentation.
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0002
EXAMPLE 5
Figure imgf000061_0001
5'-me yl-6-(3-methyl-1H-pyrazol-1-yl)-3,3'-bipyridine-5-carboxylic acid (5-2) To a solution of methyl 2-chloro-5-iodonicotonate (5J_, 10.85 g, 36.5 mmol) in dimethylformamide (150 mL) at 25°C was added 3-methyl-5-pyridylboronic acid (5.0 g, 36.5 mmol), PdCl2dppf (2.67 g, 3.65 mmol) followed by cesium carbonate (41.6 g, 128 mmol) and water (6.57 mL, 365 mmol) and the system was stirred for 4h at 25°C. The system was partitioned between water and EtOAc, and dried over magnesium sulfate. Filtration and concentration yielded a brown oil which upon purification via normal phase chromatography (0— »100% EtOAc in Hx) afforded a brown semi-solid which was then tritirated with MeOH and diethylether to yield a dark tan powder. To this tan powder (0.5 g, 1.9 mmol) in dioxane (13 mL) was added 3-methylpyrazole (0.47 g, 5.7 mmol) and NaHMDS (1.9 mL, 3.81 mmol) and the system was heated to 125°C for 20 minutes in the microwave reactor. The reaction contents were partitioned between water and EtOAc followed by purification via normal phase chromatography (20→ 100% EtOAc in Hx) to yield a clear oil. To this clear oil (0.31 g, 1.0 mmol) in THF (2.5 mL) and MeOH (2.5 mL) was added KOH (2.0 mL, 2.0 mmol) and stirred at 135°C for 10 minutes in a microwave reactor. The system was then acidified using 6 N HCl to a pH of 2.0 and the solvents were azeotroped off with toluene to afford the title compound (5-2) as a bone powder. ESI+ MS [M+H]+ C16H14N4O2 = 295.1.
5'-methyl-6-(3-methyl-1H -pyrazol-1-yl)-N-|-(l -methyl- 1,2, 3,4-tetrahvdroquinolin-6-yl)methyll- 3,3'-bipyridine-5-carboxamide (5-3) To a solution of 5'-methyl-6-(3-methyl-1H-pyrazol-1-yl)-3,3'-bipyridine-5- carboxylic acid (5-2, 0.025 g, 0.085 mmol) and l-(l-methyl-1,2,3,4-tetrahydroquinolin-6- yl)methanamine (0.015 g, 0.085 mmol) in ACN (0.85 mL) was added 1 -propylphosphonic acid cyclic anhydride (T3P, 0.15 mL, 0.255 mmol) followed by Hunig's base (0.089 mL, 0.51 mmol) and the system was stirred at 40°C for Ih in an oil bath. The reaction mixture was cooled and partitioned between EtOAc and water, washed with saturated sodium bicarbonate and dried over magnesium sulfate. The reaction mixture was filtered, concentrated and purified via normal phase chromatography (O→ 10% MeOH in EtOAc) to afford the title compound (5-3) as a bone powder. 1H NMR (500 MHz, CDCl3) δ 8.68 (m, 1H), 8.51 (m, 1H), 8.29 (m, 1H), 8.22 (m, 1H), 7.72 (m, 1H), 7.12 (m, 1H), 7.0 (m, 1H), 6.90 (m, 1H), 6.54 (m, 1H), 6.27 (m, 1H), 4.46 (m, 2H), 3.22(m, 2H), 2.88 (s, 3H), 2.74 (m, 2H), 2.44 (s, 3H), 2.28 (s, 3H), 1.97 (m, 2H). HRMS [M+H] C27H28N6O calc'd 453.2391, found 453.2397.
TABLE 5 The following compounds were prepared using the foregoing methodology, but substituting the appropriately substituted reagent, as described in the foregoing Reaction Schemes and Examples. The requisite starting materials were commercially available, described in the literature or readily synthesized by one skilled in the art of organic synthesis without undue experimentation.
Figure imgf000063_0001
Figure imgf000064_0002
Figure imgf000064_0001
Methyl 6-cvclopentyl-5'-methyl-3,3'-bipyridine-5-carboxylate (6-2)
To a solution of methyl 6-chloro-5'-methyl-3,3'-bipyridine-5-carboxylate (6-1. 0.15 g, 0.57 mmol, 1.0 equiv, prepared in an analogous fashion to Examples 2 and 8) in dioxane (2.9 mL) at 25°C was added cyclopentylzinc bromide (3.43 niL, 1.71 mmol, 3.0 equiv, 0.5 M in THF) and tetrakis(triphenylphosphine)palladium(0) (0.099 g, 0.086 mmol, 0.15 equiv). The reaction mixture was heated for 3 h at 90°C. The reaction mixture was cooled partitioned between water (10 mL) and EtOAc (30 mL), washed with water (3 x 30 mL) and brine (1 x 30 mL), and the organic phase was dried over magnesium sulfate. The residue was purified via normal phase chromatography (5 to 100% EtOAc in hexanes, silica) to afford the desired product (6-2) as an oil after concentration. ESI+ MS [M+H]+ C18H20N2O2 calc'd 297.2, found 297.1.
Sodium 6-cyclopentyl-5'-methyl-3,3'-bipyridine-5-carboxylate (6-3) To a solution of methyl 6-cyclopentyl-5'-methyl-3,3'-bipyridine-5-carboxylate (6z
2, 0.060 g, 0.020 mmol, 1.0 equiv) in THF/methanol (1.7 mL / 0.3 mL) was added 10 N sodium hydroxide (0.10 mL, 1.0 mmol, 5.0 equiv) and the system was stirred for 18 h at ambient temperature. The reaction mixture was concentrated and azeotroped with ethyl acetate (2 x 10 mL) and toluene (3 x 10 mL) to afford the desired product (6-3) as a white solid. ESI+ MS [M+H]+ C17H18N2O2 calc'd 283.1 , found 283.0.
6-cvclopentyl-N-[(5,6-dimethoxypyridin-2-yl)methyl]-5'-methyl-3,3'-bipyridine-5-carboxamide (6-4)
To a suspension of sodium 6-cyclopentyl-5'-methyl-3,3'-bipyridine-5-carboxylate (6-3, 0.022 g, 0.078 mmol, 1.0 equiv) in acetonitrile (0.8 mL) was added l-(5,6- dimethoxypyridin-2-yl)methanamine (1 1-6, 0.039 g, 0.234 mmol, 3.0 equiv), 1- propylphosphonic acid cyclic anhydride (0.149 g, 0.234 mmol, 3.0 equiv, 50 weight percent in EtOAc), and diisopropylethylamine (0.060 g, 0.47 mmol, 6.0 equiv) and the system was heated to 40°C for 24 h. The reaction mixture was cooled and diluted with ethyl acetate (30 mL). The reaction mixture was washed with saturated sodium bicarbonate (2 x 10 mL), water (2 x 10 mL) and brine (1 x 10 mL), dried over magnesium sulfate and concentrated. The residue was purified via normal phase chromatography (0 to 40% methanol in EtOAc, silica) followed by reverse phase chromatography (5 to 65% acetonitrile in water, 0.1% TFA buffer) to afford the desired product (6^4) as a white solid after free-basing and concentration. 1H NMR (500 MHz, CDCl3) δ 8.83 (d, J= 2.5Hz, 1H), 8.62 (d, J= 2.0Hz, 1H), 8.47 (s, 1H), 7.84 (d, J= 2.0Hz, 1H), 7.65 (s,
1H), 7.05 (d, J= 8.0Hz, 1H), 6.90 (d, J= 7.5Hz, 1H), 6.72 (bs, 1H), 4.64 (d, J= 5.0Hz, 2H), 3.97 (s, 3H), 3.88 (s, 3H), 3.52 - 3.48 (m, 1H), 2.42 (s, 3H), 2.04 - 1.80 (m, 6H), 1.70 - 1.60 (m, 2H). HRMS [M+H] C25H28N4O3 calc'd 433.2234, found 433.2226. TABLE 6
The following compounds were prepared using the foregoing methodology, but substituting the appropriately substituted reagent, as described in the foregoing Reaction Schemes and Examples. The requisite starting materials were commercially available, described in the literature or readily synthesized by one skilled in the art of organic synthesis without undue experimentation.
Figure imgf000066_0002
EXAMPLE 7
Figure imgf000066_0001
7 3
5-bromo-2,3-dimethoxypyridine (7-2)
To a solution of 2,3-dimthoxypyridine (7-1, 2.5 g, 18.0 mmol, 1.0 equiv) in dichloromethane: saturated NaHCO3 (80 mL: 40 mL) at 0°C was added bromine (0.93 itiL, 18.0 mmol, 1.0 equiv) and the reaction mixture was stirred for 2 h at 25°C. The reaction mixture was quenched with solid Na2SO3 (~10 g) and the aqueous phase was extracted with dichloromethane (3 x 100 mL). The organic phase was dried over magnesium sulfate and concentrated. The residue was purified via normal phase chromatography (0 to 20% EtOAc in hexanes, silica) to afford the desired product (7-2) as an oil after concentration. ESI+ MS [M+H]+ C7H8NO2 calc'd 218.0, found 218.0.
5 ,6-dimethoxynicotinonitrile (7-3)
To a solution of 5-bromo-2,3-dimethoxypyridine (7-2, 0.300 g, 1.38 mmol, 1.0 equiv) in dimethylformamide (3.9 mL) was added copper (I) cyanide (0.15 g, 1.65 mmol, 1.2 equiv) and the reaction mixture was heated for 40 minutes at 180 °C in a microwave reactor. The reaction mixture was cooled and partitioned between EtOAc (50 mL) and water (50 mL). The organic phase was washed with water (2 x 30 mL) and brine (1 x 30 mL), dried over magnesium sulfate and concentrated. The residue was purified via normal phase chromatography (0 to 40% EtOAc in hexanes, silica) to afford the desired product (7-3) as white solid after concentration. ESI+ MS [M+H]+ C8H8N2O2 calc'd 165.1 , found 165.1.
l-(5,6-dimethoxypyridin-3-yl)methanamine (7-4)
To a solution of 5,6-dimethoxynicotinonitrile (7-3, 0.600 g, 3.65 mmol, 1.0 equiv) in methanol (20 mL) under a nitrogen atmosphere was added Pearlman's catalyst (0.205 g, 0.292 mmol, 0.08 equiv, 20 weight percent) and concentrated HCl (2.44 mL), and the reaction was place under an atmosphere of hydrogen. After 2 h, the reaction was placed under an atmosphere of nitrogen and filtered through celite to remove the catalyst. The reaction mixture was dissolved in EtOAc, dried over MgSO4 and concentrated to an oily solid (7-4) of high purity. ESI+ MS [M+H]+ C8H12N2O2 calc'd 169.1, found 169.1.
EXAMPLE 8
Figure imgf000067_0001
3-cvclopropyl-4-methoxybenzonitrile (8-2)
A mixture of 3-bromo-4-methoxybenzonitrile (8-1., 5.0g, 23.6 mmol, 1.0 eq), aqueous potassium phosphate tribasic (65.0 ml, 1.27 M, 3.5 eq), cyclopropylboronic acid (10.1 g, 1 18 mmol, 5.0 eq), Pd(OAc)2 (0.539 g, 2.36 mmol, 0.1 eq) and tricyclohexylphosphine (0.661 g, 2.36 mmol, 0.1 eq) was stirred in degassed toluene (103 ml) and heated to 80°C for three hours. An additional amount of cyclopropylboronic acid (1.Og, 1.16 mmol, 0.5 eq) was added and the solution was further heated for 16 hours at 8O0C to complete the reaction. The reaction mixture was cooled and partitioned between brine and EtOAc. The organic phase was dried over sodium sulfate, filtered and concentrated to give an orange oil. The oil was purified by normal phase column chromatography (0 to 10% EtOAc in hexanes) to afford the product (8-2) as a yellow oil. 1H NMR (CDCl3) δ 7.44 (dd, 1H, J=8.4, 2.0 Hz), 7.09 (d, 1H, J= 2.0 Hz), 6.86 (d, 1H, J= 8.4 Hz), 3.92 (s, 3H), 2.13 (m, 1H), 0.97 (m, 2H), 0.65 (m, 2H).
1 -(3-cvclopropyl-4-methoxyphenyl)methanarnine (8-3)
Lithium aluminum hydride in diethyl ether (17.3 ml, 1.0 M, 17.3 mmol, 3.0 eq) was carefully added over twenty minutes to a solution of 3-cyclopropyl-4-methoxybenzonitrile (8-2, 1.Og, 5.77 mmol, 1.0 eq) in tetrahydrofuran (28.9 ml) at 0°C under nitrogen atmosphere. The resulting dark orange mixture was allowed to stir at 0°C for 30 min, then carefully quenched in the following order: water (1.OmI), 15% NaOH aqueous (1.0 ml) and water (3.0 ml). The resulting emulsion was stirred at room temperature for 30 min. Several spatula amounts of magnesium sulfate was added to the mixture to remove any water. The entire mixture was filtered through a pad of sodium sulfate, washing with ethyl acetate. The collected filtrate was concentrated to give the product (8-3) as a yellow oil. ESI+ MS [M-16]+ C 11HI 5NO: 161.1 found, 161.2 required.
EXAMPLE 9
Figure imgf000068_0001
6-Bromo-5-methoxvpyridine-2-carbonitrile (9-2) To a solution of 2-bromo-6-iodo-3-methoxypryidine (9-1 or 1 1-3. 2.1 g, 6.69 mmol, 1.0 equiv) in dichloromethane (17 mL) was added isopropylmagnesium chloride (2.0 M, 4.35 mL, 8.70 mmol, 1.3 equiv) slowly at 0 °C and the reaction mixture was stirred for 45 minutes. The reaction mixture was then cooled to -78 °C and a solution of toluenesulfonyl cyanide (1.8 g, 10.03 mmol, 1.5 equiv) in dichloromethane (5 mL) was added slowly. The reaction mixture was allowed to warm to ambient temperature over 4 hours. To quench, 2 N HCl was added until neutral pH was obtained. The reaction mixture was partitioned between water and dichloromethane and the aqueous phase was extracted with dichloromethane (3 x 50 mL). The combined organic layer was dried over MgSO4, concentrated and the residue was purified by normal phase column chromatography (10-50% EtOAc in hexanes) to afford the product (9-2) as a tan solid. ESI+ MS [M+H]+ C7H6BrN2O: 212.9 found, 213.0 required.
5-Methoxy-6-vinylpyridine-2-carbonitrile (9-3)
In a microwave vial, 6-bromo-5-methoxypyridine-2-carbonitrile (9-2, 200 mg, 0.94 mmol, 1.0 equiv), potassium trifluoro(vinyl)borate (314 mg, 2.35 mmol, 2.5 equiv), tricyclohexylphosphine (52.7 mg, 0.19 mmol, 0.2 equiv), palladium(II) acetate (21.1 mg, 0.09 mmol, 0.1 equiv), and tripotassium phosphate (697 mg, 3.29 mmol, 3.5 equiv) were suspended in toluene (10 mL) and water (0.5 mL). The reaction mixture was heated in a microwave reactor for 20 minutes at 130°C and then filtered, rinsing with EtOAc. The filtrate was concentrated and the residue purified by normal phase column chromatography (10-35% EtOAc in hexanes) to afford the product (9-3). ESI+ MS [M+H]+ C9H9N2O: 161.0 found, 161.1 required.
1 -(6-Ethyl-5-methoxypyridin-2-yl)methanamine dihydrochloride (9-4)
To a suspension of 5-methoxy-6-vinylpyridine-2-carbonitrile (9-3, 88 mg, 0.55 mmol, 1 equiv) in methanol (3 mL) was added palladium hydroxide on carbon (19.29 mg, 0.03 mmol, 0.05 equiv) and concentrated HCl (360 uL, 4.40 mmol, 8 equiv) and the reaction mixture stirred under an atmosphere of hydrogen overnight. The reaction mixture was filtered over celite, rinsing with methanol, and concentrated to give desired product (9j4) as the dihydrochloride salt. ESI+ MS [M+H]+ C9H15N2O: 167.0 found, 167.2 required.
6-Cvclopropyl-5-methoxypyridine-2-carbonitrile (9-5)
In a microwave vial, 6-bromo-5-methoxypyridine-2-carbonitrile (9^, 300 mg, 1.41 mmol, 1.0 equiv), potassium cyclopropyl(trifluoro)borate (521 mg, 3.52 mmol, 2.5 equiv), tricyclohexylphosphine (158 mg, 0.56 mmol, 0.4 equiv), palladium(II) acetate (63.2 mg, 0.28 mmol, 0.2 equiv) and tripotassium phosphate (1046 mg, 4.93 mmol, 3.5 equiv) were suspended in toluene (15 mL) and water (0.75 mL). The reaction mixture was heated in a microwave reactor for 60 minutes at 130 °C and then filtered, rinsing with EtOAc. The filtrate was concentrated and the residue purified by normal phase column chromatography (10-35% EtOAc in hexanes) to afford the product (9-5). ESI+ MS [M+H]+ Ci0HnN2O: 175.1 found, 175.2 required.
l-(6-cvclopropyl-5-methoxypyridin-2-yl) methanamine (9-6)
To a solution of 6-cyclopropyl-5-methoxypyridine-2-carbonitrile (9-5, 1 15 mg, 0.66 mmol, 1 equiv) in THF (2.6 mL) at 0 °C was added LAH (1 M, 1.98 mL, 1.98 mmol, 3 equiv) slowly. The reaction mixture was stirred and allowed to warm to ambient temperature over 3 hours. The reaction mixture was then diluted with EtOAc (2 mL), and water (50 uL), NaOH (aq., 20 wt. %, 50 uL) and water (100 uL) were added sequentially. The reaction mixture was then dried over MgSO4, filtered and concentrated to afford the product (9^). ESI+ MS [M+H]+ C10H15N2O: 179.1 found, 179.2 required.
EXAMPLE 10
Figure imgf000070_0001
3 ,4-Bis(difluoromethoxy)benzaldehyde ( 10-2) In a sealed tube 3,4-dihydroxybenzaldehyde (10-1, 750 mg, 5.43 mmol, 1.0 equiv), sodium chlorodifluoroacetate (3310 mg, 21.7 mmol, 4 equiv), and potassium carbonate (1800 mg, 13.03 mmol, 2.4 equiv) were suspended in DMF (24.4 mL) and water (2.7 mL). The reaction mixture was stirred at 100 °C for 4 hours and was cooled to ambient temperature. HCl (6 N, 10 mL) and water (30 mL) were added and the reaction stirred for an additional 2 hours. NaOH (5 N, aq.) was added until a pH of 10 was obtained. The mixture was extracted with
MTBE (3x, 50 mL), dried over MgSO4 and concentrated to afford the desired product (10-2). 1H NMR (CDCl3, 400 MHz) δ 9.97 (s, 1H), 7.76-7.79 (m, 2H), 7.43 (d, J=8.1Hz, 1H), 6.64 (t, J=73.8 Hz, 1H), 6.60 (t, J=73.8 Hz, 1H). 1 -[3,4-Bis('difluoromethoxy)phenyl1methanamine hydrochloride (10-3)
To a solution of 3,4-bis(difluoromethoxy)benzaldehyde (10-2, 100 mg, 0.42 mmol, 1 equiv) in THF (840 uL) was added 2-methylpropane-2-sulfinamide (56 mg, 0.46 mmol, 1.1 equiv) and titanium ethoxide (440 uL, 2.10 mmol, 5.0 equiv), and the reaction mixture stirred at ambient temperature for 4.5 hours. The reaction mixture was then cooled to 0 °C and sodium borohydride (31.8 mg, 0.84 mmol, 2.0 equiv) was added portionwise and the reaction was stirred for an additional hour. Methanol (2 mL) was added slowly and the reaction mixture was partitioned between EtOAc (20 mL) and brine (20 mL) and filtered. The organic phase was dried over MgSO4 and concentrated to afford crude iV-[3,4-bis(difluoromethoxy)benzyl]-2- methylpropane-2-sulfϊnamide, which was dissolved in methanol (2.2 mL). To the solution was added HCl (2 M in ether, 655 uL, 3 equiv), and the reaction mixture stirred for 30 minutes. It was then concentrated to afford the desired product (10-3) as a white solid. ESI+ MS [M-NH2]4- C9H7F4O2: 223.0 found, 223.0 required.
EXAMPLE 1 1
NaOMe DMF
Figure imgf000071_0001
Figure imgf000071_0002
11-4 11-5 1 1-6
2-bromo-3-hydroxy-6-iodopyridine (1 1 -2)
To a solution of 2-bromo-3-hydroxypyridine (11-1, 28 g, 161 mmol) in water (360 mL) was added K2CO3 (22.24 g, 161 mmol) and I2 (20.42 g, 80 mmol). The system was stirred for 1.5 h at ambient temperature, cooled to 0°C and then treated with concentrated HCl until solids precipitated from solution (pH~ 6.0). The solids were isolated by filtration and dried to give the title compound (11-2) as a brown solid. ESI+ MS C5H3BrINO: 299.8 found, 299.9 required. 2-bromo-3-methoxy-6-iodopyridine (1 1-3)
To a solution of 2-bromo-3-hydroxy-6-iodopyridine (1 1-2. 40 g, 133 mmol) in DMF (80 ml) was added K2CO3 (16.77 g, 121 mmol) and methyl iodide (66.3 g, 467 mmol). The system was stirred for 45 minutes at 100°C, cooled to room temperature and then treated with water (650 mL) and stirred for 0.5 h. The resulting solids that precipitated from solution were isolated by filtration and dried to give the title compound (1 1-3) as a pale brown solid. ESI+ MS C6H5BrINO: 313.8 found, 313.9 required.
2,3-dimethoxy-6-iodopyridine (11-4)
To a solution of 2-bromo-3-methoxy-6-iodopyridine (1 1-3, 34 g, 162 mmol) in DMF (65 mL) was added sodium methoxide (37 mL, 162 mmol) and heated to 100 °C. The mixture was stirred for 10 minutes and partitioned between saturated NaHCO3 and DCM. The organic phase was washed with brine, dried over Na2SO4, filtered and concentrated. The crude material was purified by gradient elution on silica (0 to 20% EtOAc in hexanes) to afford the title compound (11-4) as a white powder. ESI+ MS [M+H]+ C7H8INO2: 265.8 found, 266.0 required.
2,3-dimethoxy-6-cvanopyridine (1 1-5)
To a solution of 2,3-dimethoxy-6-iodopyridine (1 1-4, 24.0 g, 91 mmol) in DMF (181 mL) was added copper cyanide (9.73 g, 109 mmol) and heated to 150 °C for 20 minutes in a microwave reactor. The mixture was partitioned between water and EtOAc. The organic phase was washed with brine, dried over Na2SO4, filtered and concentrated. The crude material was purified by gradient elution on silica gel (0 to 40% EtOAc in hexanes) to yield the desired product (U1S) as an off-white crystalline powder. ESI+ MS [M+H]+ C8H8N2O2: 165.0 found, 165.1 required.
23-dimethoxy-6-aminomethylpyridine (1 1-6)
To a solution of 2,3-dimethoxy-6-cyanopyridine (1 1-5, 5.1g, 31.1 mmol) in MeOH (260 mL) was added Pearlman's catalyst (2.18 g, 3.1 1 mmol) and concentrated HCl (20.0 mL, 249 mmol). The system was then stirred under an atmosphere of hydrogen via a balloon for 1.5 h. The reaction contents were filtered through a pad of celite and methanol was removed in vacuo. The crude mixture was then basified using saturated Na2CO3 and then extracted using 4:1 Chloroform :Ethanol. The organic phase was washed with brine, dried over Na2SO4, filtered and concentrated to yield the desired product (11-6) as a bone semi-solid. ESI+ MS [M+H]+ C8Hi2N2O2: M-16 (-NH2), 152.06 found, 152.2 required.
EXAMPLE 12
Figure imgf000073_0001
3-ethyl-4-methoxybenzonitrile (12-2)
To a solution of 3-bromo-4-methoxybenzonitrile (12-1, 0.3 g, 1.42 mmol) in DMF (14 mL) was added tetraethyltin (0.56 mL, 2.83 mmol), bis(tri-t-butylphosphine) palladium(O) (0.072 g, 0.141 mmol), and lithium chloride (0.18 g, 4.24 mmol) and the system was heated to 135°C for 30 minutes in a microwave reactor. The mixture was partitioned between saturated NaHCO3 and EtOAc. The organic phase was washed with brine, dried over Na2SO4, filtered and concentrated. The crude material was purified by gradient elution on silica gel (0 to 10% EtOAc in hexanes) to yield the desired product (12-2) as a clear oil. ESI+ MS [M+H]+ Ci0Hi iNO: 162.1 found, 162.2 required.
3-ethyl-4-methoxybenzylamine (12-3)
To a solution of 3-ethyl-4-methoxybenzonitrile (12-2, 0.12 g, 0.71 mmol) in MeOH (3.5 mL) was added Pearlman's catalyst (0.020 g, 0.036 mmol) and concentrated HCl (0.47 mL, 5.71 mmol). The system was then stirred under an atmosphere of hydrogen via a balloon overnight. The reaction contents were filtered through a pad of celite and solvents were removed in vacuo to yield the desired product (12-3) as a salmon colored crystalline solid in the form of a mono HCl salt. ESI+ MS [M+H]+ C8Hi2N2O2: M-16 (-NH2), 149.07 found, 149.2 required.
EXAMPLE 13
Figure imgf000073_0002
3-methoxy-4-ethyl-benzonitrile (13-2)
To a solution of 3-methoxy-4-hydroxybenzonitrile (13-1, 2.0 g, 13.4 mmol) in DCM (67 mL) at -78 °C was added diisopropylethylamine (3.0 mL, 17.4 mmol) followed by triflic anhydride (2.7 mL, 16.0 mmol) and stirred at -78 °C for Ih. The mixture was poured into a separatory funnel containing a few pieces of ice and then partitioned between ice water and ether. The organic phase was washed with IN HCl and then 10% Na2CO3, dried over Na2SO4, filtered and concentrated to yield a pale yellow oil. To a solution of this oil (2.7 g, 9.60 mmol) in DMF (96.0 mL) was added tetraethyl tin (3.80 mL, 19.2 mmol), bis(tri-t-butylphosphine) palladium(O) (0.491 g, 0.960 mmol) and LiCl (1.22g, 28.8 mmol) and the system was heated to 80 °C for Ih in an oil bath. The mixture was partitioned between saturated NaHCO3 and EtOAc. The organic phase was washed with water, brine, dried over Na2SO4, filtered and concentrated. The crude material was purified by gradient elution on silica gel (0 to 20% EtOAc in hexanes) to afford the desired compound (13-2) as a pale yellow crystalline solid. ESI+ MS [M+H]+ Ci0Hi iNO: 162.1 found, 162.2 required.
3-methoxy-4-ethylbenzylamine (13-3)
To a solution of 3-methoxy-4-ethyl-benzonitrile (13-2, 1.5 g, 9.31 mmol) in THF (45 mL) at 0 °C was added LAH (40 mL, 40.0 mmol) and stirred at 0 °C for 0.5h. Quenched carefully over a period of 0.5h with EtOAc at 0 °C and stirred for 15 minutes. Following this, water (1.5 mL) was added to this mixture and stirred for 15 minutes followed by 15% NaOH (1.5 mL ) and again stirred for 15 minutes. Lastly, water (4.5 mL )was added and stirred for 15 more minutes. The mixture was dried over Mg2SO4, filtered and concentrated to yield a clear semisolid. This semi-solid was treated with 2.0M HCl in ether (4.65 mL, 9.3 mmol) and the reaction contents were concentrated to afford the desired product as a white powder (13-3) in the form of a mono HCl salt. ESI+ MS [M+H]+ C8Hi2N2O2: M-16 (-NH2), 149.1 found, 149.2 required.
EXAMPLE 14
Figure imgf000074_0001
2,3-dimethoxy-5-aminomethylpyrazine ( 14-2)
To a solution of 2,3-dichloropyrazine (14-1, 3.5 g, 23.5 mmol) in MeOH (1 15 mL) was added NaOMe (15.0 mL, 70.5 mmol) and the system was stirred overnight. The reaction contents were then filtered through a fritted funnel of medium porosity, concentrated, and partitioned between EtOAc and water. The organic phase was washed with brine, dried over Na2SO4, filtered and concentrated to afford a clear oil. To this clear oil (2.5 g, 17.8 mmol) in DMF (17 ml) at 0°C was added NBS (3.5 g, 19.6 mmol) the system was stirred overnight. The system was quenched with Na2SO3 and then poured into ice water. The resulting solids that precipitated from solution were isolated by filtration and dried to afford a white solid. To this white solid (1 g, 4.5 mmol) in DMF (9 mL) was added copper cyanide (0.45 g, 5.0 mmol) and heated to 185 °C for 20 minutes in the microwave reactor. The mixture was cooled and partitioned between EtOAc and water. The organic phase was washed with brine, dried over Na2SO4, filtered and concentrated. The crude material was purified by gradient elution on silica (0 to 65% EtOAc in hexanes) to afford a white powder. To this white powder (0.740 g, 4.5 mmol) in MeOH (40 mL) was added Pearlman's catalyst (0.315 g, 0.45 mmol) and concentrated HCl (3.0 mL, 36 mmol). The system was then stirred under an atmosphere of hydrogen via a balloon for 1.5 h. The reaction contents were filtered through a pad of celite followed by removal of methanol in vacuo. The crude mixture was then dissolved in DCM, basified using saturated Na2CO3 and then extracted several times with DCM. The organic phase was washed with brine, dried over Na2SO4, filtered and concentrated to yield the desired product (14-2) as a bone solid. ESI+ MS [M+H]+ C7Hi 1N3O2: M-16 (-NH2), 152.8 found, 152.2 required.
EXAMPLE 15
Figure imgf000075_0001
6-Isopropenyl-5-methoxypyridine-2-carbonitrile (15-2)
In a microwave vial, 6-bromo-5-methoxypyridine-2-carbonitrile (15-1 or 9-2, 200 mg, 0.94 mmol, 1.0 equiv), potassium trifluoro(isopropenyl)borate (Molander, Gary A., J. Am. Chem. Soc. Commun. 2003, 125, 11148-1 1 149.) (347 mg, 2.35 mmol, 2.5 equiv), tricyclohexylphosphine (52.7 mg, 0.19 mmol, 0.2 equiv), palladium(II) acetate (21.1 mg, 0.09 mmol, 0.1 equiv), and tripotassium phosphate (697 mg, 3.29 mmol, 3.5 equiv) were suspended in toluene (10 mL) and water (0.5 mL). The reaction mixture was heated in a microwave reactor for 20 minutes at 130 °C and then filtered, rinsing with EtOAc. The filtrate was concentrated and the residue purified by normal phase column chromatography (15-40% EtOAc in hexanes) to afford the title compound (JJLl)- ESI+ MS [M+H]+ C|0HnN2O: 175.1 found, 175.2 required.
l-(6-Isopropyl-5-methoxypyridin-2-yl)methanamine dihvdrochloride (T 5-3) To a suspension of 6-isopropenyl-5-methoxypyridine-2-carbonitrile (15-2, 132 mg, 0.76 mmol, 1 equiv) in methanol (4 mL) was added palladium hydroxide on carbon (26.6 mg, 0.04 mmol, 0.05 equiv) and concentrated HCl (500 uL, 6.06 mmol, 8 equiv) and the reaction mixture stirred under an atmosphere of hydrogen for three hours. The reaction mixture was filtered over celite, rinsing with methanol, and concentrated to give the title compound (15-3) as the dihydrochloride salt. ESI+ MS [M+H]+ Ci0Hi7N2O: 181.1 found, 181.3 required.
EXAMPLE 16
Figure imgf000076_0001
2-Ethoxy-6-iodo-3-methoxypyridine (16-2) In a microwave vial, 2-bromo-6-iodo-3-methoxypyridine (16-1 or 131-3, 150 mg,
0.48 mmol, 1 equiv) was dissolved in DMF (300 uL). Sodium ethoxide (268 uL, 21 wt% in EtOH, 0.72 mmol, 1.5 equiv) was added to the solution, and the reaction was heated in the microwave for 5 minutes at 100°C. The reaction mixture was partitioned between saturated NaHCO3 and DCM. The organic phase was washed once with brine, dried over Mg2SO4 and concentrated. The residue was purified by normal phase column chromatography (0-20% EtOAc in hexanes) to give the title compound (16-2). ESI+ MS [M+H]+ C8HnINO2: 280.0 found, 280.1 required.
6-Ethoxy-5-methoxypyridine-2-carbonitrile (16-3) To a solution of 2-ethoxy-6-iodo-3-methoxypyridine (16-2, 100 mg, 0.36 mmol, 1 equiv) in DMF (675 uL) was added copper cyanide (39 mg, 0.43, mmol, 1.2 equiv). The reaction mixture was heated in a microwave reactor at 150°C for 20 minutes, and then was partitioned between DCM (5OmL) and water (50 mL). The aqueous layer was extracted with DCM (3 x 25 mL), and the organic phase was dried over Mg2SO4, concentrated and purified by normal phase column chromatography (20-50% EtOAc in hexanes) to yield the title compound (16-3) as a white solid. ESI+ MS [M+H]+ C9HuN2O2: 179.1 found, 179.2 required.
l-(6-Ethoxy-5-methoxypyridin-2-yl)methanamine dihvdrochloride (16-4)
To a solution of 6-ethoxy-5-methoxypyridine-2-carbonitrile (16-3, 48 mg, 0.27 mmol, 1 equiv) in methanol (1.5 mL) was added 20 weight percent palladium hydroxide on carbon (9.5 mg, 0.01 mmol, 0.05 equiv) and concentrated HCl (177 uL, 2.16 mmol, 8 equiv) and the reaction mixture stirred under an atmosphere of hydrogen overnight. The reaction mixture was filtered over celite, rinsing with methanol, and concentrated to give the title compound (16- 4) as the dihydrochloride salt. ESI+ MS [M+H]+ C9H15N2O2: 183.1 found, 183.2 required.
TABLE
The following table shows representative data for the compounds of the Examples as orexin receptor antagonists as determined by the foregoing assays.
Figure imgf000077_0001
Figure imgf000078_0001
While the invention has been described and illustrated with reference to certain particular embodiments thereof, those skilled in the art will appreciate that various adaptations, changes, modifications, substitutions, deletions, or additions of procedures and protocols may be made without departing from the spirit and scope of the invention.

Claims

WHAT IS CLAIMED IS:
1. A compound of the formula I:
Figure imgf000079_0001
wherein: A1 is selected from the group consisting of phenyl, naphthyl and heteroaryl;
A2 is selected from the group consisting of phenyl, naphthyl and heteroaryl;
A3 is selected from the group consisting of phenyl, naphthyl, C3-6cycloalkyl, and heterocycle;
RIa, R1 b and R1° may be absent if the valency of A1 does not permit such substitution and are independently selected from the group consisting of:
(1) hydrogen,
(2) halogen,
(3) hydroxyl,
(4) -(C=O)m-On-C1 -6alkyl, where m is 0 or 1, n is 0 or 1 (wherein if m is 0 or n is 0, a bond is present) and where the alkyl is unsubstituted or substituted with one or more substituents selected from R13,
(5) -(C=O)m-On-C3-6cycloalkyl, where the cycloalkyl is unsubstituted or substituted with one or more substituents selected from R13 ;
(6) -(C=O)m-C2-4alkenyl, where the alkenyl is unsubstituted or substituted with one or more substituents selected from R13,
(7) -(C=O)m-C2-4alkynyl, where the alkynyl is unsubstituted or substituted with one or more substituents selected from R13, (8) -(C=O)m-On-phenyl or -(C=O)m-On-napthyl, where the phenyl or naphthyl is unsubstituted or substituted with one or more substituents selected from R13,
(9) -(C=O)m-On-heterocycle, where the heterocycle is unsubstituted or substituted with one or more substituents selected from R13, (10) -(C=0)m-NR10 R 1 1, wherein R10 and R 1 1 are independently selected from the group consisting of:
(a) hydrogen,
(b) C1 -6alkyl, which is unsubstituted or substituted with R13,
(c) C3-6alkenyl, which is unsubstituted or substituted with R13 s (d) C3-6alkynyl, which is unsubstituted or substituted with R13,
(e) C3-6cycloalkyl which is unsubstituted or substituted with R13,
(f) phenyl, which is unsubstituted or substituted with R13, and
(g) heterocycle, which is unsubstituted or substituted with R13, (1 1) -S(0)2-NR1θR1 l, (12) -S(O)q-R12, where q is 0, 1 or 2 and where R12 is selected from the definitions of R1O and R1 I,
(13) -CO2H,
(14) -CN, and
(15) -NO2;
R2a, R2b and R2c may be absent if the valency of A2 does not permit such substitution and are independently selected from the group consisting of:
(1) hydrogen,
(2) halogen, (3) hydroxyl,
(4) -(C=O)m-On-C1 -6alkyl, where the alkyl is unsubstituted or substituted with one or more substituents selected from R13,
(5) -(C=O)m-On-C3-6cycloalkyl, where the cycloalkyl is unsubstituted or substituted with one or more substituents selected from R13, (6) -(C=O)m-C2-4alkenyl, where the alkenyl is unsubstituted or substituted with one or more substituents selected from R13,
(7) -(C=O)m-C2-4alkynyl, where the alkynyl is unsubstituted or substituted with one or more substituents selected from R13,
(8) -(C=O)m-On-phenyl or -(C=O)m-On-napthyl, where the phenyl or naphthyl is unsubstituted or substituted with one or more substituents selected from R13,
(9) -(C=O)m-On-heterocycle, where the heterocycle is unsubstituted or substituted with one or more substituents selected from R13, (10) -(C=O)1n-NR10R1 1,
(1 1) -S(0)2-NR10R1 1,
(12) -S(O)q-R12,
(13) -CO2H, (14) -CN, and
(15) -NO2;
R3a ; R3b and R3c may be absent if the valency of A3 does not permit such substitution and are independently selected from the group consisting of: (1) hydrogen,
(2) halogen,
(3) hydroxyl,
(4) -(C=O)m-On-C 1 -6alkyl, where the alkyl is unsubstituted or substituted with one or more substituents selected from R13, (5) -(C=O)m-On-C3-6cycloalkyl, where the cycloalkyl is unsubstituted or substituted with one or more substituents selected from R13,
(6) -(C=O)m-C2-4alkenyl, where the alkenyl is unsubstituted or substituted with one or more substituents selected from R13,
(7) -(C=O)m-C2-4alkynyl, where the alkynyl is unsubstituted or substituted with one or more substituents selected from R 13,
(8) -(C=O)m-On-phenyl or -(C=O)m-On-napthyl, where the phenyl or naphthyl is unsubstituted or substituted with one or more substituents selected from R13,
(9) -(C=O)m-On-heterocycle, where the heterocycle is unsubstituted or substituted with one or more substituents selected from R13, (10) -(C=O)1n-NR10R1 1,
(1 1) -S(O)2-NR10R1 1 ,
(12) -S(0)q-R12,
(13) -CO2H,
(14) -CN, and (15) -NO2; with the proviso that if A3 is pyridyl, at least one of R3a, R3b and R3c is other than hydrogen;
R4 and R5 are independently selected from hydrogen and C1 -6alkyl, which is unsubstituted or substituted with one or more substituents selected from R13, or R4 and R5 may be joined together to form a C3-6cycloalkyl with the carbon atom to which they are attached, where the cycloalkyl is unsubstituted or substituted with one or more substituents selected from R13; R6 is hydrogen, C1 -6alkyl or C3-6cycloalkyl, which is unsubstituted or substituted with one or more substituents selected from R13;
R13 is selected from the group consisting of: (1) halogen,
(2) hydroxyl,
(3) -(C=O)m-On-C 1 -6alkyl, where the alkyl is unsubstituted or substituted with one or more substituents selected from R 14,
(4) -On-(C1 -3)perfluoroalkyl, (5) -(C=O)m-On-C3-6cycloalkyl, where the cycloalkyl is unsubstituted or substituted with one or more substituents selected from R 14,
(6) -(C=O)m-C2-4alkenyl, where the alkenyl is unsubstituted or substituted with one or more substituents selected from R 14,
(7) -(C=O)m-C2-4alkynyl, where the alkynyl is unsubstituted or substituted with one or more substituents selected from R 14,
(8) -(C=O)m -On-phenyl or -(C=O)m-On-napthyl, where the phenyl or naphthyl is unsubstituted or substituted with one or more substituents selected from R14,
(9) -(C=O)m-On-heterocycle, where the heterocycle is unsubstituted or substituted with one or more substituents selected from R 14, (10) -(C=O)1n-NR10R1 1,
(1 1) -S(0)2-NR1θR1 l,
(12) -S(O)q-R12,
(13) -CO2H,
(14) -CN, and (15) -NO2;
R14 is selected from the group consisting of:
(1) hydroxyl,
(2) halogen, (3) C1 -6alkyl,
(4) -Cβ-όcycloalkyl,
(5) -O-C1 -6alkyl,
(6) -0(C=O)-C l-6alkyl,
(7) -NH-C1 -6alkyl, (8) phenyl,
(9) heterocycle,
(10) -CO2H, and (H) -CN; or a pharmaceutically acceptable salt thereof.
2. The compound of Claim 1 wherein A1 is phenyl.
3. The compound of Claim 1 wherein A1 is pyridyl.
4. The compound of Claim 1 wherein A2 is phenyl.
5. The compound of Claim 1 wherein A2 is heteroaryl.
6. The compound of Claim 1 wherein A3 is phenyl.
7. The compound of Claim 1 wherein R 1 a, R1 b and R1 c are independently selected from the group consisting of:
(1) hydrogen,
(2) halogen,
(3) hydroxyl,
(4) C1 -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl, phenyl or napthyl,
(5) -O-C1 -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl or phenyl,
(6) heteroaryl, wherein heteroaryl is selected from pyrrolyl, imidazolyl, indolyl, pyridyl, and pyrimidinyl, which is unsubstituted or substituted with halogen, hydroxyl, C 1 -6alkyl, -O-C 1 -6alkyl or-NO2,
(7) phenyl, which is unsubstituted or substituted with halogen, hydroxyl, C1 -6alkyl, -O-C1 -6alkyl or-NO2,
(8) -O-phenyl, which is unsubstituted or substituted with halogen, hydroxyl, C1 -6alkyl, -O-C1 -6alkyl or-NO2, and (9) -NH-C l -6alkyl, or -N(C 1 -6alkyl)(C I -6alkyl), which is unsubstituted or substituted with halogen, hydroxyl, C1 -6alkyl, -O-C1 -6alkyl or-NO2-
8. The compound of Claim 7 wherein R 1 a, R1 b and R 1 c are independently selected from the group consisting of: (1) hydrogen,
(2) halogen,
(3) hydroxyl, (4) C1 -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl or phenyl or napthyl, and
(5) -O-C1 -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl or phenyl.
9. The compound of Claim 8 wherein R 1 a, R1 b and R1 c are independently selected from the group consisting of:
(1) hydrogen,
(2) chloro, (3) fluroro, and
(4) methyl.
10. The compound of Claim 1 wherein R2a, R2b and R2c are independently selected from the group consisting of: (1) hydrogen,
(2) halogen,
(3) hydroxyl,
(4) C1 -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl or phenyl or napthyl, (5) -O-C1 -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl or phenyl,
(6) heteroaryl, wherein heteroaryl is selected from pyrrolyl, imidazolyl, indolyl, pyridyl, and pyrimidinyl, which is unsubstituted or substituted with halogen, hydroxyl, C1 -6alkyl, -O-C1 -6alkyl or-NO2, (7) phenyl, which is unsubstituted or substituted with halogen, hydroxyl, C1 -6alkyl, -
O-C1 -6alkyl or-NO2,
(8) -O-phenyl, which is unsubstituted or substituted with halogen, hydroxyl, C l . 6alkyl, -O-C1 -6alkyl or-NO2, and
(9) -NH-C 1 -6alkyl, or -N(C I -6alkyl)(C I -6alkyl), which is unsubstituted or substituted with halogen, hydroxyl, C I -6alkyl, -O-C 1 -6alkyl or-NO2-
1 1. The compound of Claim 10 wherein R2a ; R2b and R2c are independently selected from the group consisting of:
(1) hydrogen, (2) chloro,
(3) fluoro,
(4) bromo, (5) methoxy,
(6) t-butoxy,
(7) difluoromethyl, and
(8) trifluoromethyl,
(9) -N(CH3).
12. The compound of Claim 1 wherein R3a, R3b and R3c are independently selected from the group consisting of:
(1) hydrogen, (2) halogen,
(3) hydroxyl,
(4) C1 -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl, phenyl or napthyl,
(5) -O-C1 -6alkyl, which is unsubstituted or substituted with halogen, hydroxyl or phenyl,
(6) heteroaryl, wherein heteroaryl is selected from pyrrolyl, imidazolyl, indolyl, pyridyl, and pyrimidinyl, which is unsubstituted or substituted with halogen, hydroxyl, C1 -6alkyl, -O-C1 -6alkyl or-NO2,
(7) phenyl, which is unsubstituted or substituted with halogen, hydroxyl, C1 -6alkyl, -O-C1 -6alkyl or-NO2,
(8) -O-phenyl, which is unsubstituted or substituted with halogen, hydroxyl, C]-6alkyl, -O-C1 -6alkyl or-NO2, and
(9) -NH-C 1 -6alkyl, or -N(C 1 -6alkyl)(C I -6alkyl), which is unsubstituted or substituted with halogen, hydroxyl, C1 -6alkyl, -O-C1 -6alkyl or-NO2; with the proviso that if A3 is pyridyl, at least one of R3a, R3b and R3c is other than hydrogen.
13. The compound of Claim 12 wherein R3a ? R3b and R3c are independently selected from the group consisting of:
(1) hydrogen, (2) halogen, and
(3) C1 -6alkyl; with the proviso that if A3 is pyridyl, at least one of R3a ; R3b and R3c is other than hydrogen.
14. A compound which is selected from the group consisting of: N-(3,4-Dimethoxybenzyl)-5-(3,5-dimethylphenyl)-2-(l-methyl-1H-pyrazol-4-yl)nicotinamide; N-(3)4.dimethoxybenzyl)-5-(3,5-dimethylphenyl)-6'-fluoro-2,3'-bipyridine-3-carboxamide; N-(3,4-dimethoxybenzyl)-5-(3,5-dimethylphenyl)-2-quinolin-3-ylnicotinamide; N-(3,4-dimethoxybenzyl)-5-(3,5-dimethylphenyl)-2-(3-hydroxyphenyl)nicotinamide;
N-CS^-dimethoxybenzyl)-5-CS^-dimethylphenyl)^-IS-tCmethylamino^arbony^phenyl}- nicotinamide;
N-(3 ,4-dimethoxybenzyl)-2- { 3-[(dimethylamino)methyl] phenyl } -5-(3 ,5-dimethylphenyl) nicotinamide;
N-(3,4-dimethoxybenzyl)-5-(3,5-dimethylphenyl)-2-(1H-indol-5-yl)nicotinamide;
5 -(3 ,5 -dimethylphenyl)-N- [( 1 R)- 1 -(3 -methoxyphenyl)ethyl] -2-( 1 -methyl- 1 H-pyrazol-4- yl)nicotinamide;
5-(3, 5-dimethylphenyl)-2-(l -methyl- 1H-pyrazol-4-yl)-N-(quinolin-2-ylmethyl) nicotinamide; N-[(5,6-dimethoxypyridin-3-yl)methyl]-5-(3,5-dimethylphenyl)-2-(l -methyl- 1H-pyrazol-4- yl)nicotinamide;
5 -(3 ,5 -dichlorophenyl)-N- [(2 ,3 -dimethyl- 1 H-indol-6-yl)methyl] -2-( 1 -methyl- 1 H-pyrazol-4- yl)nicotinamide;
5-(3-fluoro-5-methylphenyl)-2-(l-methyl-1H-pyrazol-4-yl)-N-[(lR)-1-(2- naphthyl)ethyl] nicotinamide;
5-(3,5-dimethylphenyl)-2-(l -methyl- 1H-pyrazol-4-yl)-N-(2-naphthylmethyl) nicotinamide;
5-(3-fluoro-5-methylphenyl)-2-(l -methyl- 1H-pyrazol-4-yl)-N-(2-naphthylmethyl) nicotinamide;
5-(3,5-dimethylphenyl)-2-(l -methyl- 1H-pyrazol-4-yl)-N-(quinolin-3-ylmethyl)nicotinamide;
5-(3-Chloro-5-methylphenyl)-N-(3,4-dimethoxybenzyl)-2-(l-methyl-1H-pyrazol-4-yl) nicotinamide;
5-(3 ,5 -fluoromethylphenyl)-N- [ 1 -(3 ,4-dimethoxyphenyl) ethyl] -2-( 1 -methyl- 1 H-pyrazol-4- yl)nicotinamide;
5-(3,5-fluoromethylphenyl)-N-(3,4-dimethoxybenzyl)-2-(l-methyl-1H-pyrazol-4- yl)nicotinamide; 5-(3,5-chloromethylphenyl)-N-[(2,3-dimethyl-1-H-indol-5-yl)methyl]-2-(l -methyl- 1H-pyrazol-4- yl)nicotinamide;
5-(3,5-chloromethylphenyl)-N-(2-naphthylmethyl)-2-(l -methyl- 1H-pyrazol-4-yl)nicotinamide;
5-(3,5-fluoromethylphenyl)-N-(3,4-dimethoxybenzyl)-2-pyridazin-3yl-nicotinamide;
5-(3,5-chloromethylphenyl)-N-(3,4-dichlorobenzyl)-2-(l-methyl-1H-pyrazol-4-yl)nicotinamide; 5-(3,5-fluoromethylphenyl)-N-[ 1 -(3 ,4-dimethoxyphenyl) ethyl]-6'-fluoro-2,3'-bipyridine-3- carboxamide;
5-(3,5-dichlorophenyl)-N-(3,4-dimethoxybenzyl)-5'-chloro-2,3'-bipyridine-3-carboxamide;
5-(3,5-chloromethylphenyl)-N-[(l-methyl-1H-benzimidazol-2-yl) methyl]-2-(l-methyl-1H- pyrazol-4-yl)nicotinamide; 5-(3,5-chloromethylphenyl)-N-[( 1 -methyl-2,3-dihydro- 1 H-indol-5-yl) methyl]-2-( 1 -methyl- 1 H- pyrazol-4-yl)nicotinamide; 5-(3,5-chloromethylphenyl)-N-[(1,4-dimethyl-1,2,3,4-tetrahydroquinoxalin-6-yl) methyl]-2-(1- methyl- 1 H-pyrazol-4-yl)nicotinamide; 5-(3,5-chloromethylphenyl)-N-[(1,4,5-trimethyl-1H-imidazol-2-yl)methyl]-2-(1-methyl-1H- pyrazol-4-yl)nicotinamide; 5-(3,5-fluoromethylphenyl)-N-(3,4-dimethoxybenzyl)-2-(l-methyl-1H-pyrazol-5- yl)nicotinamide;
5-(3,5-chloromethylphenyl)-N-(3,4-dihydroxybenzyl)-2-(l-methyl-1H-pyrazol-4- yl)nicotinamide;
5-(3,5-chloromethylphenyl)-N-[( 1 -methyl- 1 H-indol-2-yl)methyl] -2-( 1 -methyl- 1 H-pyrazol-4- yl)nicotinamide;
N-[(5,6-dimethoxypyridin-2-yl)methyl]-6-(2-fluorophenyl)-5'-methyl-3,3'-bipyridine-5- carboxamide;
N-[(5,6-dimethoxypyridin-2-yl)methyl]-6-(3-fluorophenyl)-5'-methyl-3,3'-bipyridine-5- carboxamide; 6-(3 -chlorophenyl)-N- [(5 ,6-dimethoxypyridin-2-yl)methyl] -5 '-methyl-3 ,3 '-bipyridine-5- carboxamide;
6-(3-chlorophenyl)-N-[(5,6-dimethoxypyridin-3-yl)methyl]-5'-methyl-3,3'-bipyridine-5- carboxamide;
N- [(5 ,6-dimethoxypyridin-2-yl)methyl] -5 '-methyl-6-phenyl-3 ,3 '-bipyridine-5 -carboxamide ; N-^-chloro^-methoxybenzyl)-5'-methyl-6-phenyl-3,3'-bipyridine-5-carboxamide;
N-[(5,6-dimethoxypyrazin-2-yl)methyl]-6-(2-fluorophenyl)-5'-methyl-3,3'-bipyridine-5- carboxamide;
N-(3-chloro-4-methoxybenzyl)-5'-methyl-6-(3-methylphenyl)-3,3'-bipyridine-5-carboxamide;
6-(2,3 -difluorophenyl)-N- [(5 ,6-dimethoxypyridin-2-yl)methyl] -5 '-methyl-3 ,3 '-bipyridine-5 - carboxamide;
5'-chloro-N-[(5,6-dimethoxypyridin-2-yl)methyl]-6-phenyl-3,3'-bipyridine-5-carboxamide;
N-[(6-cyclopropyl-5-methoxypyridin-2-yl)methyl]-6-(3-fluorophenyl)-5'-methyl-3,3'-bipyridine-
5-carboxamide;
6-(3,5-difluorophenyl)-N-[(5,6-dimethoxypyridin-2-yl)methyl]-5'-methyl-3,3'-bipyridine-5- carboxamide;
6-(3,5-difluorophenyl)-N-[(5,6-dimethoxypyrazin-2-yl)methyl]-5'-methyl-3,3'-bipyridine-5- carboxamide;
6-(3-chloro-5-fluorophenyl)-N-[(5,6-dimethoxypyridin-2-yl)methyl]-5'-methyl-3,3'-bipyridine-5- carboxamide; 6-(3-cyanophenyl)-N-[(5,6-dimethoxypyridin-2-yl)methyl]-5'-methyl-3,3'-bipyridine-5- carboxamide;
5-(3-chloro-5-methylphenyl)-N-(3,4-dimethoxybenzyl)-2-(morpholin-4-yl)nicotinamide; 5-(3-chloro-5-methylphenyl)-N-(3,4-dimethoxybenzyl)-2-piperidin-1-ylnicotinamide;
5-(3-chloro-5-methylphenyl)-N-(3,4-dimethoxybenzyl)-2-pyrrolidin-1-ylnicotinamide;
2-azetidin-1-yl-5-(3-chloro-5-methylphenyl)-N-(3,4-dimethoxybenzyl)-nicotinamide;
5-(3,5-dichlorophenyl)-N-(3,4-dimethoxybenzyl)-2-(3-methoxyazetidin-1-yl)nicotinamide; 5-(3,5-dichlorophenyl)-N-(3,4-dimethoxybenzyl)-2-(3-fluoroazetidin-1-yl)nicotinamide;
5-(3-chloro-5-methylphenyl)-N-(3,4-dimethoxybenzyl)-2-[(3R)-3-fluoropyrrolidin-1- yl]nicotinamide;
5-(3-chloro-5-methylphenyl)-N-(3,4-dimethoxybenzyl)-2-thiomorpholin-4-ylnicotinamide;
5-(3,5-dichlorophenyl)-N-[(5,6-dimethoxypyridin-2-yl)methyl]-2-morpholin-4-ylnicotinamide; 5 '-chloro-N- [(5 ,6-dimethoxypyridin-2-yl)methyl] -6-morpholin-4-yl-3 ,3 '-bipyridine-5 - carboxamide;
N- [(5 ,6-dimethoxypyridin-2-yl)m ethyl] -5 '-methyl-6-morpholin-4-yl-3 ,3 '-bipyridine-5 - carboxamide;
5-(3-chloro-5-methylphenyl)-N-[(5,6-dimethoxypyrazin-2-yl)methyl]-2-morpholin-4- ylnicotinamide;
N-[4-methoxy-3-(trifluoromethyl)benzyl]-5'-methyl-6-pyrrolidin-1-yl-3,3'-bipyridine-5- carboxamide;
N-(3 -chloro-4-methoxybenzyl)-5 '-methyl-6-pyrrolidin- 1 -yl-3 ,3 '-bipyridine-5 -carboxamide ;
5'-methyl-6-(3-methyl- 1 H -pyrazol- 1 -yl)-N-[( 1 -methyl- 1 ,2,3 ,4-tetrahydroquinolin-6-yl)methyl] - 3,3'-bipyridine-5-carboxamide;
N-[(5,6-dimethoxypyridin-2-yl)methyl]-5-(3,5-dimethylphenyl)-2-(4-methyl-1H-pyrazol-1- yl)nicotinamide;
5-(3-chloro-5-fluorophenyl)-N-[(5,6-dimethoxypyridin-2-yl)methyl]-2-(4-methyl-1H-pyrazol-1- yl)nicotinamide; N-(3-cyclopropyl-4-methoxybenzyl)-5'-methyl-6-(3-methyl- 1 H-pyrazol- 1 -yl)-3,3'-bipyridine-5- carboxamide;
N-(3-chloro-4-methoxybenzyl)-5-(3-fluoro-5-methoxyphenyl)-2-(1H-pyrazol-1-yl)nicotinamide;
N-(3-chloro-4-methoxybenzyl)-5'-methyl-6-(l H-pyrazol- l-yl)-3,3'-bipyridine-5-carboxamide;
N- [(5 ,6-dimethoxypyridin-2-yl)methyl] -5 '-methyl-6-(3 -methyl- 1 H-pyrazol- 1 -yl)-3 ,3 '-bipyridine- 5-carboxamide;
N-(3-ethyl-4-methoxybenzyl)-5'-methyl-6-(3-methyl-l H-pyrazol- l-yl)-3,3'-bipyridine-5- carboxamide;
N-(3-chloro-4-methoxybenzyl)-5'-methyl-6-(3-methyl-l H-pyrazol- l-yl)-3,3'-bipyridine-5- carboxamide; 5-(3-chloro-5-methylphenyl)-N-(3,4-dimethoxybenzyl)-2-(4-methyl-l H-pyrazol- 1- yl)nicotinamide;
6-cyclopentyl-N-[(5,6-dimethoxypyridin-2-yl)methyl]-5'-methyl-3,3'-bipyridine-5-carboxamide; 6-cyclopentyl-N-(3,4-dimethoxybenzyl)-5'-methyl-3,3'-bipyridine-5-carboxamide; N-(3-chloro-4-methoxybenzyl)-6-cyclopentyl-5'-methyl-3,3'-bipyridine-5-carboxamide; 6-cyclopentyl-N-[4-methoxy-3-(trifluoromethyl)benzyl]-5'-methyl-3,3'-bipyridine-5- carboxamide; or a pharmaceutically acceptable salt thereof.
15. A pharmaceutical composition which comprises an inert carrier and a compound of Claim 1 or a pharmaceutically acceptable salt thereof.
16. A compound of Claim 1 or a pharmaceutically acceptable salt thereof for use in medicine.
17. Use of a compound of Claim 1, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment or prevention of a sleep disorder.
18. A method for enhancing the quality of sleep in a mammalian patient in need thereof which comprises administering to the patient a therapeutically effective amount of the compound of Claim 1 or a pharmaceutically acceptable salt thereof.
19. A method for treating insomnia in a mammalian patient in need thereof which comprises administering to the patient a therapeutically effective amount of the compound of Claim 1 or a pharmaceutically acceptable salt thereof.
20. A method for treating or controlling obesity in a mammalian patient in need thereof which comprises administering to the patient a therapeutically effective amount of the compound of Claim 1 or a pharmaceutically acceptable salt thereof.
PCT/US2008/009491 2007-08-09 2008-08-07 Pyridine carboxamide orexin receptor antagonists WO2009020642A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
JP2010519985A JP2010535769A (en) 2007-08-09 2008-08-07 Pyridine carboxamide orexin receptor antagonist
CA2695621A CA2695621A1 (en) 2007-08-09 2008-08-07 Pyridine carboxamide orexin receptor antagonists
US12/669,909 US8003797B2 (en) 2007-08-09 2008-08-07 Pyridine carboxamide orexin receptor antagonists
EP08795115A EP2184981A4 (en) 2007-08-09 2008-08-07 Pyridine carboxamide orexin receptor antagonists
AU2008284268A AU2008284268A1 (en) 2007-08-09 2008-08-07 Pyridine carboxamide orexin receptor antagonists
US13/197,800 US20110301140A1 (en) 2007-08-09 2011-08-04 Pyridine Carboxamide orexin Receptor Antagonists

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US96411107P 2007-08-09 2007-08-09
US60/964,111 2007-08-09

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/197,800 Division US20110301140A1 (en) 2007-08-09 2011-08-04 Pyridine Carboxamide orexin Receptor Antagonists

Publications (1)

Publication Number Publication Date
WO2009020642A1 true WO2009020642A1 (en) 2009-02-12

Family

ID=40341600

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/009491 WO2009020642A1 (en) 2007-08-09 2008-08-07 Pyridine carboxamide orexin receptor antagonists

Country Status (6)

Country Link
US (2) US8003797B2 (en)
EP (1) EP2184981A4 (en)
JP (1) JP2010535769A (en)
AU (1) AU2008284268A1 (en)
CA (1) CA2695621A1 (en)
WO (1) WO2009020642A1 (en)

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010051236A1 (en) * 2008-10-30 2010-05-06 Merck Sharp & Dohme Corp. Isonicotinamide orexin receptor antagonists
WO2010141275A1 (en) * 2009-06-01 2010-12-09 Merck Sharp & Dohme Corp. Pyrazine carboxamide orexin receptor antagonists
EP2349267A1 (en) * 2008-10-30 2011-08-03 Merck Sharp & Dohme Corp. Pyridazine carboxamide orexin receptor antagonists
US8546403B2 (en) 2010-04-27 2013-10-01 Calcimedica, Inc. Compounds that modulate intracellular calcium
US8618307B2 (en) 2009-09-16 2013-12-31 Calcimedica, Inc. Compounds that modulate intracellular calcium
US8754219B2 (en) 2010-04-27 2014-06-17 Calcimedica, Inc. Compounds that modulate intracellular calcium
CN104350053A (en) * 2012-06-15 2015-02-11 大正制药株式会社 Heteroaromatic methyl cyclic amine derivative
CN104364238A (en) * 2012-06-15 2015-02-18 大正制药株式会社 Branched chain alkyl heteroaromatic ring derivative
US9079891B2 (en) 2010-08-27 2015-07-14 Calcimedica, Inc. Compounds that modulate intracellular calcium
US9440982B2 (en) 2012-02-07 2016-09-13 Eolas Therapeutics, Inc. Substituted prolines/piperidines as orexin receptor antagonists
US9499517B2 (en) 2012-02-07 2016-11-22 Eolas Therapeutics, Inc. Substituted prolines / piperidines as orexin receptor antagonists
WO2017194548A1 (en) 2016-05-10 2017-11-16 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for the treatment of autoimmune inflammatory diseases
US9856240B2 (en) 2011-10-19 2018-01-02 Calcimedica, Inc. Compounds that modulate intracellular calcium
US20180235959A1 (en) 2015-08-07 2018-08-23 Calcimedica, Inc. Use of crac channel inhibitors for the treatment of stroke and traumatic brain injury
US10106529B2 (en) 2011-06-10 2018-10-23 Calcimedia, Inc. Compounds that modulate intracellular calcium
US10221170B2 (en) 2014-08-13 2019-03-05 Eolas Therapeutics, Inc. Difluoropyrrolidines as orexin receptor modulators
US10703722B2 (en) 2010-04-27 2020-07-07 Calcimedica, Inc. Compounds that modulate intracellular calcium
US10821109B1 (en) 2015-02-27 2020-11-03 Calcimedica, Inc. Pyrazine-containing compound
US10894789B2 (en) 2016-02-12 2021-01-19 Astrazeneca Ab Halo-substituted piperidines as orexin receptor modulators
US11596633B2 (en) 2017-09-07 2023-03-07 Revolution Medicines, Inc. SHP2 inhibitor compositions and methods for treating cancer
US11661401B2 (en) 2016-07-12 2023-05-30 Revolution Medicines, Inc. 2,5-disubstituted 3-methyl pyrazines and 2,5,6-trisubstituted 3-methyl pyrazines as allosteric SHP2 inhibitors
US11673901B2 (en) 2017-12-15 2023-06-13 Revolution Medicines, Inc. Polycyclic compounds as allosteric SHP2 inhibitors
US11673896B2 (en) 2017-01-23 2023-06-13 Revolution Medicines, Inc. Pyridine compounds as allosteric SHP2 inhibitors
US11702411B2 (en) 2017-10-12 2023-07-18 Revolution Medicines, Inc. Pyridine, pyrazine, and triazine compounds as allosteric SHP2 inhibitors
US11739093B2 (en) 2017-01-23 2023-08-29 Revolution Medicines, Inc. Substituted pyrazolopyrazines, imidazopyrazines and [1,2,4]triazolopyrazines as allosteric SHP2 inhibitors

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AR072899A1 (en) 2008-08-07 2010-09-29 Merck Sharp & Dohme DERIVATIVES OF TERPIRIDINE-CARBOXAMIDE ANTAGONISTS OF OREXIN RECEPTORS, PHARMACEUTICAL COMPOSITIONS CONTAINING THEM AND USE OF THE SAME IN THE TREATMENT OF INSOMNIUM AND OBESITY.
WO2010051237A1 (en) * 2008-10-30 2010-05-06 Merck Sharp & Dohme Corp. 2,5-disubstituted phenyl carboxamide orexin receptor antagonists
EP2763974B1 (en) * 2011-10-05 2016-09-14 Merck Sharp & Dohme Corp. Phenyl carboxamide-containing spleen tyrosine kinase (syk) inhibitors
US9216173B2 (en) 2011-10-05 2015-12-22 Merck Sharp & Dohme Corp. 2-Pyridyl carboxamide-containing spleen tyrosine kinase (SYK) inhibitors
EP2763975B1 (en) * 2011-10-05 2016-04-06 Merck Sharp & Dohme Corp. 3-pyridyl carboxamide-containing spleen tyrosine kinase (syk) inhibitors
AR088352A1 (en) * 2011-10-19 2014-05-28 Merck Sharp & Dohme ANTAGONISTS OF THE RECEIVER OF 2-PIRIDILOXI-4-NITRILE OREXINE
US8883857B2 (en) 2012-12-07 2014-11-11 Baylor College Of Medicine Small molecule xanthine oxidase inhibitors and methods of use
JP5930010B2 (en) * 2013-12-13 2016-06-08 大正製薬株式会社 Medicament containing heteroaromatic methyl cyclic amine derivative
TWI649307B (en) * 2014-05-07 2019-02-01 日商橘生藥品工業股份有限公司 Cyclohexylpyridine derivative
JP6986955B2 (en) * 2016-12-28 2021-12-22 キッセイ薬品工業株式会社 Method for producing pyrazole derivative

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020147198A1 (en) * 2001-01-12 2002-10-10 Guoqing Chen Substituted arylamine derivatives and methods of use
WO2007061763A2 (en) * 2005-11-22 2007-05-31 Merck & Co., Inc. Indole orexin receptor antagonists

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0126292D0 (en) * 2001-11-01 2002-01-02 Smithkline Beecham Plc Compounds
GB0130341D0 (en) * 2001-12-19 2002-02-06 Smithkline Beecham Plc Compounds
EP1871752A4 (en) * 2005-04-12 2009-09-30 Merck & Co Inc Amidopropoxyphenyl orexin receptor antagonists
US8410142B2 (en) * 2007-03-02 2013-04-02 Merck Sharp & Dohme Corp. Bipyridine carboxamide orexin receptor antagonists
AR072899A1 (en) * 2008-08-07 2010-09-29 Merck Sharp & Dohme DERIVATIVES OF TERPIRIDINE-CARBOXAMIDE ANTAGONISTS OF OREXIN RECEPTORS, PHARMACEUTICAL COMPOSITIONS CONTAINING THEM AND USE OF THE SAME IN THE TREATMENT OF INSOMNIUM AND OBESITY.
PE20110852A1 (en) 2008-10-30 2011-11-25 Merck Sharp & Dohme ISONICOTINAMIDE OREXIN RECEPTOR ANTAGONISTS

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020147198A1 (en) * 2001-01-12 2002-10-10 Guoqing Chen Substituted arylamine derivatives and methods of use
WO2007061763A2 (en) * 2005-11-22 2007-05-31 Merck & Co., Inc. Indole orexin receptor antagonists

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP2184981A4 *

Cited By (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2349267A1 (en) * 2008-10-30 2011-08-03 Merck Sharp & Dohme Corp. Pyridazine carboxamide orexin receptor antagonists
CN102272103A (en) * 2008-10-30 2011-12-07 默沙东公司 Isonicotinamide orexin receptor antagonists
EP2349267A4 (en) * 2008-10-30 2012-08-29 Merck Sharp & Dohme Pyridazine carboxamide orexin receptor antagonists
WO2010051236A1 (en) * 2008-10-30 2010-05-06 Merck Sharp & Dohme Corp. Isonicotinamide orexin receptor antagonists
US8592457B2 (en) 2008-10-30 2013-11-26 Merck Sharp & Dohme Corp. Isonicotinamide orexin receptor antagonists
WO2010141275A1 (en) * 2009-06-01 2010-12-09 Merck Sharp & Dohme Corp. Pyrazine carboxamide orexin receptor antagonists
US8618307B2 (en) 2009-09-16 2013-12-31 Calcimedica, Inc. Compounds that modulate intracellular calcium
US10703722B2 (en) 2010-04-27 2020-07-07 Calcimedica, Inc. Compounds that modulate intracellular calcium
US9120751B2 (en) 2010-04-27 2015-09-01 Calcimedica, Inc. Compounds that modulate intracellular calcium
US11905248B2 (en) 2010-04-27 2024-02-20 Calcimedica, Inc. Compounds that modulate intracellular calcium
US8546403B2 (en) 2010-04-27 2013-10-01 Calcimedica, Inc. Compounds that modulate intracellular calcium
US8980629B2 (en) 2010-04-27 2015-03-17 Calcimedica, Inc. Compounds that modulate intracellular calcium
TWI574959B (en) * 2010-04-27 2017-03-21 鈣系醫藥公司 Compounds that modulate intracellular calcium
US9090612B2 (en) 2010-04-27 2015-07-28 Calcimedica, Inc. Compounds that modulate intracellular calcium
US8754219B2 (en) 2010-04-27 2014-06-17 Calcimedica, Inc. Compounds that modulate intracellular calcium
US9353099B2 (en) 2010-04-27 2016-05-31 Calcimedica, Inc. Compounds that modulate intracellular calcium
US10336738B2 (en) 2010-08-27 2019-07-02 Calcimedica, Inc. Compounds that modulate intracellular calcium
US9079891B2 (en) 2010-08-27 2015-07-14 Calcimedica, Inc. Compounds that modulate intracellular calcium
US10106529B2 (en) 2011-06-10 2018-10-23 Calcimedia, Inc. Compounds that modulate intracellular calcium
US9856240B2 (en) 2011-10-19 2018-01-02 Calcimedica, Inc. Compounds that modulate intracellular calcium
US9440982B2 (en) 2012-02-07 2016-09-13 Eolas Therapeutics, Inc. Substituted prolines/piperidines as orexin receptor antagonists
US9499517B2 (en) 2012-02-07 2016-11-22 Eolas Therapeutics, Inc. Substituted prolines / piperidines as orexin receptor antagonists
US9896452B2 (en) 2012-02-07 2018-02-20 Eolas Therapeutics, Inc. Substituted prolines/piperidines as orexin receptor antagonists
RU2639869C2 (en) * 2012-06-15 2017-12-25 Тайсо Фармасьютикал Ко., Лтд. Hetero-aromatic methylic derivative of cyclic amine
CN104350053A (en) * 2012-06-15 2015-02-11 大正制药株式会社 Heteroaromatic methyl cyclic amine derivative
CN104350053B (en) * 2012-06-15 2017-03-08 大正制药株式会社 Heteroaromatic methyl Cycloamine derivative
US9266870B2 (en) 2012-06-15 2016-02-23 Taisho Pharmaceutical Co., Ltd Heteroaromatic methyl cyclic amine derivative
CN104364238A (en) * 2012-06-15 2015-02-18 大正制药株式会社 Branched chain alkyl heteroaromatic ring derivative
US10221170B2 (en) 2014-08-13 2019-03-05 Eolas Therapeutics, Inc. Difluoropyrrolidines as orexin receptor modulators
US11311535B2 (en) 2015-02-27 2022-04-26 Calcimedica, Inc. Pancreatitis treatment
US11752148B2 (en) 2015-02-27 2023-09-12 Calcimedica, Inc. Pyrazine-containing compound
US10821109B1 (en) 2015-02-27 2020-11-03 Calcimedica, Inc. Pyrazine-containing compound
US11439639B2 (en) 2015-02-27 2022-09-13 Calcimedica, Inc. Pyrazine-containing compound
US11013737B2 (en) 2015-02-27 2021-05-25 Calcimedia, Inc. Pyrazine-containing compound
US10478435B2 (en) 2015-08-07 2019-11-19 Calcimedica, Inc. Use of CRAC channel inhibitors for the treatment of stroke and traumatic brain injury
US20180235959A1 (en) 2015-08-07 2018-08-23 Calcimedica, Inc. Use of crac channel inhibitors for the treatment of stroke and traumatic brain injury
US11434236B2 (en) 2016-02-12 2022-09-06 Astrazeneca Ab Halo-substituted piperidines as orexin receptor modulators
US10894789B2 (en) 2016-02-12 2021-01-19 Astrazeneca Ab Halo-substituted piperidines as orexin receptor modulators
WO2017194548A1 (en) 2016-05-10 2017-11-16 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for the treatment of autoimmune inflammatory diseases
US11661401B2 (en) 2016-07-12 2023-05-30 Revolution Medicines, Inc. 2,5-disubstituted 3-methyl pyrazines and 2,5,6-trisubstituted 3-methyl pyrazines as allosteric SHP2 inhibitors
US11673896B2 (en) 2017-01-23 2023-06-13 Revolution Medicines, Inc. Pyridine compounds as allosteric SHP2 inhibitors
US11739093B2 (en) 2017-01-23 2023-08-29 Revolution Medicines, Inc. Substituted pyrazolopyrazines, imidazopyrazines and [1,2,4]triazolopyrazines as allosteric SHP2 inhibitors
US11596633B2 (en) 2017-09-07 2023-03-07 Revolution Medicines, Inc. SHP2 inhibitor compositions and methods for treating cancer
US11702411B2 (en) 2017-10-12 2023-07-18 Revolution Medicines, Inc. Pyridine, pyrazine, and triazine compounds as allosteric SHP2 inhibitors
US11673901B2 (en) 2017-12-15 2023-06-13 Revolution Medicines, Inc. Polycyclic compounds as allosteric SHP2 inhibitors

Also Published As

Publication number Publication date
CA2695621A1 (en) 2009-02-12
EP2184981A4 (en) 2012-02-01
US20100197652A1 (en) 2010-08-05
US8003797B2 (en) 2011-08-23
JP2010535769A (en) 2010-11-25
US20110301140A1 (en) 2011-12-08
EP2184981A1 (en) 2010-05-19
AU2008284268A1 (en) 2009-02-12

Similar Documents

Publication Publication Date Title
US8003797B2 (en) Pyridine carboxamide orexin receptor antagonists
EP2323985B1 (en) Tripyridyl carboxamide orexin receptor antagonists
EP2349262B1 (en) 2,5-disubstituted phenyl carboxamide orexin receptor antagonists
EP2350010B1 (en) Isonicotinamide orexin receptor antagonists
EP2349267B1 (en) Pyridazine carboxamide orexin receptor antagonists
US8710076B2 (en) 2,5-disubstituted piperidine orexin receptor antagonists
WO2016100154A1 (en) Pyrrolidine orexin receptor antagonists
EP2438041A1 (en) Pyrazine carboxamide orexin receptor antagonists
WO2016069519A1 (en) Pyrazole orexin receptor antagonists
WO2011005636A1 (en) Tetrahydronapthyridine orexin receptor antagonists
EP2771346A1 (en) Isoxazolopyridine orexin receptor antagonists

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08795115

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2008284268

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 12669909

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2008284268

Country of ref document: AU

Date of ref document: 20080807

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2695621

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2010519985

Country of ref document: JP

REEP Request for entry into the european phase

Ref document number: 2008795115

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2008795115

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE